메뉴 건너뛰기




Volumn , Issue 8 MAY, 2014, Pages

Advances in non-dopaminergic treatments for Parkinson's disease

Author keywords

Animal models; Dopamine; Dyskinesias; Gene therapy; L Dopa; Neurodegeneration; Parkinson's disease; Therapeutics

Indexed keywords

2 DIPROPYLAMINO 8 HYDROXYTETRALIN; 5 AMINO 2 (2 FURYL) 7 (2 PHENYLETHYL)PYRAZOLO[4,3 E][1,2,4]TRIAZOLO[1,5 C]PYRIMIDINE; 5 PROPOXY 3 (1,2,5,6 TETRAHYDRO 4 PYRIDYL)PYRROLO[3,2 B]PYRIDINE; 8 (3,4 DIMETHOXYSTYRYL) 7 METHYL 1,3 DIPROPYLXANTHINE; ADENOSINE RECEPTOR BLOCKING AGENT; ADRENERGIC RECEPTOR BLOCKING AGENT; AMPA RECEPTOR ANTAGONIST; ANTIINFLAMMATORY AGENT; ANTIPARKINSON AGENT; AROMATIC LEVO AMINO ACID DECARBOXYLASE; CALCIUM CHANNEL BLOCKING AGENT; CHELATING AGENT; GLIAL CELL LINE DERIVED NEUROTROPHIC FACTOR; GLUCAGON LIKE PEPTIDE 1 RECEPTOR AGONIST; GLUTAMATE DECARBOXYLASE; ISTRADEFYLLINE; MAVOGLURANT; METABOTROPIC RECEPTOR ANTAGONIST; N METHYL DEXTRO ASPARTIC ACID RECEPTOR BLOCKING AGENT; NEURTURIN; SEROTONIN AGONIST; TRAXOPRODIL;

EID: 84904959625     PISSN: 16624548     EISSN: 1662453X     Source Type: Journal    
DOI: 10.3389/fnins.2014.00113     Document Type: Review
Times cited : (80)

References (329)
  • 1
    • 0032837272 scopus 로고    scopus 로고
    • Range of neuropsychiatric disturbances in patients with Parkinson's disease
    • doi: 10.1136/jnnp.67.4.492
    • Aarsland, D., Larsen, J. P., Lim, N. G., Janvin, C., Karlsen, K., Tandberg, E., et al. (1999). Range of neuropsychiatric disturbances in patients with Parkinson's disease. J. Neurol. Neurosurg. Psychiatry 67, 492-496. doi: 10.1136/jnnp.67.4.492.
    • (1999) J. Neurol. Neurosurg. Psychiatry , vol.67 , pp. 492-496
    • Aarsland, D.1    Larsen, J.P.2    Lim, N.G.3    Janvin, C.4    Karlsen, K.5    Tandberg, E.6
  • 2
    • 67649607263 scopus 로고    scopus 로고
    • Activin A is essential for neurogenesis following neurodegeneration
    • doi: 10.1002/stem.80
    • Abdipranoto-Cowley, A., Park, J. S., Croucher, D., Daniel, J., Henshall, S., Galbraith, S., et al. (2009). Activin A is essential for neurogenesis following neurodegeneration. Stem Cells 27, 1330-1346. doi: 10.1002/stem.80.
    • (2009) Stem Cells , vol.27 , pp. 1330-1346
    • Abdipranoto-Cowley, A.1    Park, J.S.2    Croucher, D.3    Daniel, J.4    Henshall, S.5    Galbraith, S.6
  • 3
    • 0024450903 scopus 로고
    • The functional anatomy of basal ganglia disorders
    • doi: 10.1016/0166-2236(89)90074-X
    • Albin, R. L., Young, A. B., and Penney, J. B. (1989). The functional anatomy of basal ganglia disorders. Trends Neurosci. 12, 366-375. doi: 10.1016/0166-2236(89)90074-X.
    • (1989) Trends Neurosci. , vol.12 , pp. 366-375
    • Albin, R.L.1    Young, A.B.2    Penney, J.B.3
  • 4
    • 0028839005 scopus 로고
    • The functional anatomy of disorders of the basal ganglia
    • doi: 10.1016/0166-2236(95)80020-3
    • Albin, R. L., Young, A. B., and Penney, J. B. (1995). The functional anatomy of disorders of the basal ganglia. Trends Neurosci. 18, 63-64. doi: 10.1016/0166-2236(95)80020-3.
    • (1995) Trends Neurosci. , vol.18 , pp. 63-64
    • Albin, R.L.1    Young, A.B.2    Penney, J.B.3
  • 5
    • 0033816145 scopus 로고    scopus 로고
    • A multicenter, randomized clinical study to evaluate the effect on cognitive function of topiramate compared with valproate as add-on therapy to carbamazepine in patients with partial-onset seizures
    • doi: 10.1111/j.1528-1157.2000.tb00322.x
    • Aldenkamp, A. P., Baker, G., Mulder, O. G., Chadwick, D., Cooper, P., Doelman, J., et al. (2000). A multicenter, randomized clinical study to evaluate the effect on cognitive function of topiramate compared with valproate as add-on therapy to carbamazepine in patients with partial-onset seizures. Epilepsia 41, 1167-1178. doi: 10.1111/j.1528-1157.2000.tb00322.x.
    • (2000) Epilepsia , vol.41 , pp. 1167-1178
    • Aldenkamp, A.P.1    Baker, G.2    Mulder, O.G.3    Chadwick, D.4    Cooper, P.5    Doelman, J.6
  • 6
    • 30944435647 scopus 로고    scopus 로고
    • Following Complete Review of Phase 2 Trial Data Amgen Confirms Decision to Halt GDNF Study; Comprehensive Review of Scientific Findings, Patient Safety, Drove Decision
    • Amgen, Thousand Oaks: AMGEN.
    • Amgen. (2005). Following Complete Review of Phase 2 Trial Data Amgen Confirms Decision to Halt GDNF Study; Comprehensive Review of Scientific Findings, Patient Safety, Drove Decision. Thousand Oaks: AMGEN.
    • (2005)
  • 7
    • 73949090103 scopus 로고    scopus 로고
    • Inflammation in neurodegenerative diseases
    • doi: 10.1111/j.1365-2567.2009.03225.x
    • Amor, S., Puentes, F., Baker, D., and Van Der Valk, P. (2010). Inflammation in neurodegenerative diseases. Immunology 129, 154-169. doi: 10.1111/j.1365-2567.2009.03225.x.
    • (2010) Immunology , vol.129 , pp. 154-169
    • Amor, S.1    Puentes, F.2    Baker, D.3    Van Der Valk, P.4
  • 8
    • 0028984349 scopus 로고
    • L-DOPA is converted to dopamine in serotonergic fibers of the striatum of the rat: a double-labeling immunofluorescence study
    • doi: 10.1016/0304-3940(95)11817-G
    • Arai, R., Karasawa, N., Geffard, M., and Nagatsu, I. (1995). L-DOPA is converted to dopamine in serotonergic fibers of the striatum of the rat: a double-labeling immunofluorescence study. Neurosci. Lett. 195, 195-198. doi: 10.1016/0304-3940(95)11817-G.
    • (1995) Neurosci. Lett. , vol.195 , pp. 195-198
    • Arai, R.1    Karasawa, N.2    Geffard, M.3    Nagatsu, I.4
  • 9
    • 33645090418 scopus 로고    scopus 로고
    • Prolonged blockade of NMDA or mGluR5 glutamate receptors reduces nigrostriatal degeneration while inducing selective metabolic changes in the basal ganglia circuitry in a rodent model of Parkinson's disease
    • doi: 10.1016/j.nbd.2005.09.010
    • Armentero, M. T., Fancellu, R., Nappi, G., Bramanti, P., and Blandini, F. (2006). Prolonged blockade of NMDA or mGluR5 glutamate receptors reduces nigrostriatal degeneration while inducing selective metabolic changes in the basal ganglia circuitry in a rodent model of Parkinson's disease. Neurobiol. Dis. 22, 1-9. doi: 10.1016/j.nbd.2005.09.010.
    • (2006) Neurobiol. Dis. , vol.22 , pp. 1-9
    • Armentero, M.T.1    Fancellu, R.2    Nappi, G.3    Bramanti, P.4    Blandini, F.5
  • 10
    • 34447647661 scopus 로고    scopus 로고
    • Common anti-inflammatory drugs are potentially therapeutic for Parkinson's disease?
    • doi: 10.1016/j.expneurol.2007.05.006
    • Asanuma, M., and Miyazaki, I. (2007). Common anti-inflammatory drugs are potentially therapeutic for Parkinson's disease? Exp. Neurol. 206, 172-178. doi: 10.1016/j.expneurol.2007.05.006.
    • (2007) Exp. Neurol. , vol.206 , pp. 172-178
    • Asanuma, M.1    Miyazaki, I.2
  • 11
    • 47749147444 scopus 로고    scopus 로고
    • The dorsal motor nucleus of the vagus is not an obligatory trigger site of Parkinson's disease
    • doi: 10.1111/j.1365-2990.2008.00937.x
    • Attems, J., and Jellinger, K. A. (2008). The dorsal motor nucleus of the vagus is not an obligatory trigger site of Parkinson's disease. Neuropathol. Appl. Neurobiol. 34, 466-467. doi: 10.1111/j.1365-2990.2008.00937.x.
    • (2008) Neuropathol. Appl. Neurobiol. , vol.34 , pp. 466-467
    • Attems, J.1    Jellinger, K.A.2
  • 12
    • 0031661173 scopus 로고    scopus 로고
    • Aspirin and salicylate protect against MPTP-induced dopamine depletion in mice
    • doi: 10.1046/j.1471-4159.1998.71041635.x
    • Aubin, N., Curet, O., Deffois, A., and Carter, C. (1998). Aspirin and salicylate protect against MPTP-induced dopamine depletion in mice. J. Neurochem. 71, 1635-1642. doi: 10.1046/j.1471-4159.1998.71041635.x.
    • (1998) J. Neurochem. , vol.71 , pp. 1635-1642
    • Aubin, N.1    Curet, O.2    Deffois, A.3    Carter, C.4
  • 14
    • 0031930771 scopus 로고    scopus 로고
    • Glial pathology but absence of apoptotic nigral neurons in long-standing Parkinson's disease
    • doi: 10.1002/mds.870130205
    • Banati, R. B., Daniel, S. E., and Blunt, S. B. (1998). Glial pathology but absence of apoptotic nigral neurons in long-standing Parkinson's disease. Mov. Disord. 13, 221-227. doi: 10.1002/mds.870130205.
    • (1998) Mov. Disord. , vol.13 , pp. 221-227
    • Banati, R.B.1    Daniel, S.E.2    Blunt, S.B.3
  • 15
    • 26444586231 scopus 로고    scopus 로고
    • Effects of serotonin 5-HT1A agonist in advanced Parkinson's disease
    • doi: 10.1002/mds.20370
    • Bara-Jimenez, W., Bibbiani, F., Morris, M. J., Dimitrova, T., Sherzai, A., Mouradian, M. M., et al. (2005). Effects of serotonin 5-HT1A agonist in advanced Parkinson's disease. Mov. Disord. 20, 932-936. doi: 10.1002/mds.20370.
    • (2005) Mov. Disord. , vol.20 , pp. 932-936
    • Bara-Jimenez, W.1    Bibbiani, F.2    Morris, M.J.3    Dimitrova, T.4    Sherzai, A.5    Mouradian, M.M.6
  • 16
    • 0042626108 scopus 로고    scopus 로고
    • Adenosine A(2A) receptor antagonist treatment of Parkinson's disease
    • doi: 10.1212/01.WNL.0000073136.00548.D4
    • Bara-Jimenez, W., Sherzai, A., Dimitrova, T., Favit, A., Bibbiani, F., Gillespie, M., et al. (2003). Adenosine A(2A) receptor antagonist treatment of Parkinson's disease. Neurology 61, 293-296. doi: 10.1212/01.WNL.0000073136.00548.D4.
    • (2003) Neurology , vol.61 , pp. 293-296
    • Bara-Jimenez, W.1    Sherzai, A.2    Dimitrova, T.3    Favit, A.4    Bibbiani, F.5    Gillespie, M.6
  • 17
    • 79959977446 scopus 로고    scopus 로고
    • IFN-gamma signaling, with the synergistic contribution of TNF-alpha, mediates cell specific microglial astroglial activation in experimental models of Parkinson's disease.
    • doi: 10.1038/cddis.2011.17
    • Barcia, C., Ros, C. M., Annese, V., Gomez, A., Ros-Bernal, F., Aguado-Yera, D., et al. (2011). IFN-gamma signaling, with the synergistic contribution of TNF-alpha, mediates cell specific microglial and astroglial activation in experimental models of Parkinson's disease. Cell Death Dis. 2:e142. doi: 10.1038/cddis.2011.17.
    • (2011) Cell Death Dis. , vol.2
    • Barcia, C.1    Ros, C.M.2    Annese, V.3    Gomez, A.4    Ros-Bernal, F.5    Aguado-Yera, D.6
  • 18
    • 83555178498 scopus 로고    scopus 로고
    • Effects of noradrenergic denervation on L-DOPA-induced dyskinesia and its treatment by alpha- and beta-adrenergic receptor antagonists in hemiparkinsonian rats
    • doi: 10.1016/j.pbb.2011.09.009
    • Barnum, C. J., Bhide, N., Lindenbach, D., Surrena, M. A., Goldenberg, A. A., Tignor, S., et al. (2012). Effects of noradrenergic denervation on L-DOPA-induced dyskinesia and its treatment by alpha- and beta-adrenergic receptor antagonists in hemiparkinsonian rats. Pharmacol. Biochem. Behav. 100, 607-615. doi: 10.1016/j.pbb.2011.09.009.
    • (2012) Pharmacol. Biochem. Behav. , vol.100 , pp. 607-615
    • Barnum, C.J.1    Bhide, N.2    Lindenbach, D.3    Surrena, M.A.4    Goldenberg, A.A.5    Tignor, S.6
  • 19
    • 84879086501 scopus 로고    scopus 로고
    • Safety/feasibility of targeting the substantia nigra with AAV2-neurturin in Parkinson patients
    • doi: 10.1212/WNL.0b013e3182904faa
    • Bartus, R. T., Baumann, T. L., Siffert, J., Herzog, C. D., Alterman, R., Boulis, N., et al. (2013). Safety/feasibility of targeting the substantia nigra with AAV2-neurturin in Parkinson patients. Neurology 80, 1698-1701. doi: 10.1212/WNL.0b013e3182904faa.
    • (2013) Neurology , vol.80 , pp. 1698-1701
    • Bartus, R.T.1    Baumann, T.L.2    Siffert, J.3    Herzog, C.D.4    Alterman, R.5    Boulis, N.6
  • 20
    • 79951476995 scopus 로고    scopus 로고
    • Bioactivity of AAV2-neurturin gene therapy (CERE-120): differences between Parkinson's disease and nonhuman primate brains
    • doi: 10.1002/mds.23442
    • Bartus, R. T., Herzog, C. D., Chu, Y., Wilson, A., Brown, L., Siffert, J., et al. (2011). Bioactivity of AAV2-neurturin gene therapy (CERE-120): differences between Parkinson's disease and nonhuman primate brains. Mov. Disord. 26, 27-36. doi: 10.1002/mds.23442.
    • (2011) Mov. Disord. , vol.26 , pp. 27-36
    • Bartus, R.T.1    Herzog, C.D.2    Chu, Y.3    Wilson, A.4    Brown, L.5    Siffert, J.6
  • 21
    • 77956217986 scopus 로고    scopus 로고
    • Parkinson's disease: a model dilemma
    • doi: 10.1038/466S8a
    • Beal, M. F. (2010). Parkinson's disease: a model dilemma. Nature 466, S8-S10. doi: 10.1038/466S8a.
    • (2010) Nature , vol.466
    • Beal, M.F.1
  • 22
    • 0025980955 scopus 로고
    • The iron chelator desferrioxamine (Desferal) retards 6-hydroxydopamine-induced degeneration of nigrostriatal dopamine neurons
    • doi: 10.1111/j.1471-4159.1991.tb11444.x
    • Ben-Shachar, D., Eshel, G., Finberg, J. P., and Youdim, M. B. (1991). The iron chelator desferrioxamine (Desferal) retards 6-hydroxydopamine-induced degeneration of nigrostriatal dopamine neurons. J. Neurochem. 56, 1441-1444. doi: 10.1111/j.1471-4159.1991.tb11444.x.
    • (1991) J. Neurochem. , vol.56 , pp. 1441-1444
    • Ben-Shachar, D.1    Eshel, G.2    Finberg, J.P.3    Youdim, M.B.4
  • 23
    • 0026692807 scopus 로고
    • Role of iron and iron chelation in dopaminergic-induced neurodegeneration: implication for Parkinson's disease
    • doi: 10.1002/ana.410320718
    • Ben-Shachar, D., Eshel, G., Riederer, P., and Youdim, M. B. (1992). Role of iron and iron chelation in dopaminergic-induced neurodegeneration: implication for Parkinson's disease. Ann. Neurol. 32(Suppl.), S105-S110. doi: 10.1002/ana.410320718.
    • (1992) Ann. Neurol. , vol.32 , Issue.SUPPL.
    • Ben-Shachar, D.1    Eshel, G.2    Riederer, P.3    Youdim, M.B.4
  • 24
    • 79959372108 scopus 로고    scopus 로고
    • AFQ056 treatment of levodopa-induced dyskinesias: results of 2 randomized controlled trials
    • doi: 10.1002/mds.23616
    • Berg, D., Godau, J., Trenkwalder, C., Eggert, K., Csoti, I., Storch, A., et al. (2011). AFQ056 treatment of levodopa-induced dyskinesias: results of 2 randomized controlled trials. Mov. Disord. 26, 1243-1250. doi: 10.1002/mds.23616.
    • (2011) Mov. Disord. , vol.26 , pp. 1243-1250
    • Berg, D.1    Godau, J.2    Trenkwalder, C.3    Eggert, K.4    Csoti, I.5    Storch, A.6
  • 25
    • 33750320296 scopus 로고    scopus 로고
    • Iron metabolism in Parkinsonian syndromes
    • doi: 10.1002/mds.21020
    • Berg, D., and Hochstrasser, H. (2006). Iron metabolism in Parkinsonian syndromes. Mov. Disord. 21, 1299-1310. doi: 10.1002/mds.21020.
    • (2006) Mov. Disord. , vol.21 , pp. 1299-1310
    • Berg, D.1    Hochstrasser, H.2
  • 26
    • 84881556666 scopus 로고    scopus 로고
    • Study of the antidyskinetic effect of eltoprazine in animal models of levodopa-induced dyskinesia
    • doi: 10.1002/mds.25366
    • Bezard, E., Tronci, E., Pioli, E. Y., Li, Q., Porras, G., Bjorklund, A., et al. (2013). Study of the antidyskinetic effect of eltoprazine in animal models of levodopa-induced dyskinesia. Mov. Disord. 28, 1088-1096. doi: 10.1002/mds.25366.
    • (2013) Mov. Disord. , vol.28 , pp. 1088-1096
    • Bezard, E.1    Tronci, E.2    Pioli, E.Y.3    Li, Q.4    Porras, G.5    Bjorklund, A.6
  • 27
    • 33750430870 scopus 로고    scopus 로고
    • Involvement of adenosinergic receptor system in an animal model of tardive dyskinesia and associated behavioural, biochemical and neurochemical changes
    • doi: 10.1016/j.ejphar.2006.09.010
    • Bishnoi, M., Chopra, K., and Kulkarni, S. K. (2006). Involvement of adenosinergic receptor system in an animal model of tardive dyskinesia and associated behavioural, biochemical and neurochemical changes. Eur. J. Pharmacol. 552, 55-66. doi: 10.1016/j.ejphar.2006.09.010.
    • (2006) Eur. J. Pharmacol. , vol.552 , pp. 55-66
    • Bishnoi, M.1    Chopra, K.2    Kulkarni, S.K.3
  • 28
    • 34250737857 scopus 로고    scopus 로고
    • Theophylline, adenosine receptor antagonist prevents behavioral, biochemical and neurochemical changes associated with an animal model of tardive dyskinesia
    • Bishnoi, M., Chopra, K., and Kulkarni, S. K. (2007). Theophylline, adenosine receptor antagonist prevents behavioral, biochemical and neurochemical changes associated with an animal model of tardive dyskinesia. Pharmacol. Rep. 59, 181-191..
    • (2007) Pharmacol. Rep. , vol.59 , pp. 181-191
    • Bishnoi, M.1    Chopra, K.2    Kulkarni, S.K.3
  • 29
    • 0032797570 scopus 로고    scopus 로고
    • Differing effects of N-methyl-D-aspartate receptor subtype selective antagonists on dyskinesias in levodopa-treated 1-methyl-4-phenyl-tetrahydropyridine monkeys
    • Blanchet, P. J., Konitsiotis, S., Whittemore, E. R., Zhou, Z. L., Woodward, R. M., and Chase, T. N. (1999). Differing effects of N-methyl-D-aspartate receptor subtype selective antagonists on dyskinesias in levodopa-treated 1-methyl-4-phenyl-tetrahydropyridine monkeys. J. Pharmacol. Exp. Ther. 290, 1034-1040..
    • (1999) J. Pharmacol. Exp. Ther. , vol.290 , pp. 1034-1040
    • Blanchet, P.J.1    Konitsiotis, S.2    Whittemore, E.R.3    Zhou, Z.L.4    Woodward, R.M.5    Chase, T.N.6
  • 30
    • 47049116273 scopus 로고    scopus 로고
    • The 6-hydroxydopamine model: news from the past
    • doi: 10.1016/j.parkreldis.2008.04.015
    • Blandini, F., Armentero, M. T., and Martignoni, E. (2008). The 6-hydroxydopamine model: news from the past. Parkinsonism. Relat. Disord. 14(Suppl. 2), S124-S129. doi: 10.1016/j.parkreldis.2008.04.015.
    • (2008) Parkinsonism. Relat. Disord. , vol.14 , Issue.SUPPL. 2
    • Blandini, F.1    Armentero, M.T.2    Martignoni, E.3
  • 31
    • 0034287012 scopus 로고    scopus 로고
    • Functional changes of the basal ganglia circuitry in Parkinson's disease
    • doi: 10.1016/S0301-0082(99)00067-2
    • Blandini, F., Nappi, G., Tassorelli, C., and Martignoni, E. (2000). Functional changes of the basal ganglia circuitry in Parkinson's disease. Prog. Neurobiol. 62, 63-88. doi: 10.1016/S0301-0082(99)00067-2.
    • (2000) Prog. Neurobiol. , vol.62 , pp. 63-88
    • Blandini, F.1    Nappi, G.2    Tassorelli, C.3    Martignoni, E.4
  • 32
    • 0032171185 scopus 로고    scopus 로고
    • L-Type calcium channels mediate a slow excitatory synaptic transmission in rat midbrain dopaminergic neurons
    • Bonci, A., Grillner, P., Mercuri, N. B., and Bernardi, G. (1998). L-Type calcium channels mediate a slow excitatory synaptic transmission in rat midbrain dopaminergic neurons. J. Neurosci. 18, 6693-6703..
    • (1998) J. Neurosci. , vol.18 , pp. 6693-6703
    • Bonci, A.1    Grillner, P.2    Mercuri, N.B.3    Bernardi, G.4
  • 33
    • 0028057911 scopus 로고
    • Buspirone in levodopa-induced dyskinesias
    • doi: 10.1097/00002826-199402000-00008
    • Bonifati, V., Fabrizio, E., Cipriani, R., Vanacore, N., and Meco, G. (1994). Buspirone in levodopa-induced dyskinesias. Clin. Neuropharmacol. 17, 73-82. doi: 10.1097/00002826-199402000-00008.
    • (1994) Clin. Neuropharmacol. , vol.17 , pp. 73-82
    • Bonifati, V.1    Fabrizio, E.2    Cipriani, R.3    Vanacore, N.4    Meco, G.5
  • 34
    • 58149333196 scopus 로고    scopus 로고
    • Behavioral recovery in MPTP-treated monkeys: neurochemical mechanisms studied by intrastriatal microdialysis
    • doi: 10.1523/JNEUROSCI.3465-08.2008
    • Boulet, S., Mounayar, S., Poupard, A., Bertrand, A., Jan, C., Pessiglione, M., et al. (2008). Behavioral recovery in MPTP-treated monkeys: neurochemical mechanisms studied by intrastriatal microdialysis. J. Neurosci. 28, 9575-9584. doi: 10.1523/JNEUROSCI.3465-08.2008.
    • (2008) J. Neurosci. , vol.28 , pp. 9575-9584
    • Boulet, S.1    Mounayar, S.2    Poupard, A.3    Bertrand, A.4    Jan, C.5    Pessiglione, M.6
  • 35
    • 0036392590 scopus 로고    scopus 로고
    • Staging of the intracerebral inclusion body pathology associated with idiopathic Parkinson's disease (preclinical and clinical stages)
    • III, doi: 10.1007/s00415-002-1301-4
    • Braak, H., Del Tredici, K., Bratzke, H., Hamm-Clement, J., Sandmann-Keil, D., and Rub, U. (2002). Staging of the intracerebral inclusion body pathology associated with idiopathic Parkinson's disease (preclinical and clinical stages). J. Neurol. 249(Suppl. 3), III, 1-5. doi: 10.1007/s00415-002-1301-4.
    • (2002) J. Neurol. , vol.249 , Issue.SUPPL. 3 , pp. 1-5
    • Braak, H.1    Del Tredici, K.2    Bratzke, H.3    Hamm-Clement, J.4    Sandmann-Keil, D.5    Rub, U.6
  • 36
    • 0037333666 scopus 로고    scopus 로고
    • Staging of brain pathology related to sporadic Parkinson's disease
    • doi: 10.1016/S0197-4580(02)00065-9
    • Braak, H., Del Tredici, K., Rub, U., De Vos, R. A., Jansen Steur, E. N., and Braak, E. (2003). Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol. Aging 24, 197-211. doi: 10.1016/S0197-4580(02)00065-9.
    • (2003) Neurobiol. Aging , vol.24 , pp. 197-211
    • Braak, H.1    Del Tredici, K.2    Rub, U.3    De Vos, R.A.4    Jansen Steur, E.N.5    Braak, E.6
  • 37
    • 5444255434 scopus 로고    scopus 로고
    • Stages in the development of Parkinson's disease-related pathology
    • doi: 10.1007/s00441-004-0956-9
    • Braak, H., Ghebremedhin, E., Rub, U., Bratzke, H., and Del Tredici, K. (2004). Stages in the development of Parkinson's disease-related pathology. Cell Tissue. Res. 318, 121-134. doi: 10.1007/s00441-004-0956-9.
    • (2004) Cell Tissue. Res. , vol.318 , pp. 121-134
    • Braak, H.1    Ghebremedhin, E.2    Rub, U.3    Bratzke, H.4    Del Tredici, K.5
  • 38
    • 0141856474 scopus 로고    scopus 로고
    • Metabotropic glutamate 5 receptor blockade alleviates akinesia by normalizing activity of selective basal-ganglia structures in parkinsonian rats
    • Breysse, N., Amalric, M., and Salin, P. (2003). Metabotropic glutamate 5 receptor blockade alleviates akinesia by normalizing activity of selective basal-ganglia structures in parkinsonian rats. J. Neurosci. 23, 8302-8309..
    • (2003) J. Neurosci. , vol.23 , pp. 8302-8309
    • Breysse, N.1    Amalric, M.2    Salin, P.3
  • 39
    • 0036662945 scopus 로고    scopus 로고
    • Chronic but not acute treatment with a metabotropic glutamate 5 receptor antagonist reverses the akinetic deficits in a rat model of parkinsonism
    • Breysse, N., Baunez, C., Spooren, W., Gasparini, F., and Amalric, M. (2002). Chronic but not acute treatment with a metabotropic glutamate 5 receptor antagonist reverses the akinetic deficits in a rat model of parkinsonism. J. Neurosci. 22, 5669-5678.
    • (2002) J. Neurosci. , vol.22 , pp. 5669-5678
    • Breysse, N.1    Baunez, C.2    Spooren, W.3    Gasparini, F.4    Amalric, M.5
  • 40
    • 0032785896 scopus 로고    scopus 로고
    • Comparison of pallidal and subthalamic nucleus deep brain stimulation for advanced Parkinson's disease: results of a randomized, blinded pilot study
    • discussion: 1382-1374
    • Burchiel, K. J., Anderson, V. C., Favre, J., and Hammerstad, J. P. (1999). Comparison of pallidal and subthalamic nucleus deep brain stimulation for advanced Parkinson's disease: results of a randomized, blinded pilot study. Neurosurgery 45, 1375-1382; discussion: 1382-1374..
    • (1999) Neurosurgery , vol.45 , pp. 1375-1382
    • Burchiel, K.J.1    Anderson, V.C.2    Favre, J.3    Hammerstad, J.P.4
  • 41
    • 0036589970 scopus 로고    scopus 로고
    • Beyond the iron mask: towards better recognition and treatment of depression associated with Parkinson's disease
    • doi: 10.1002/mds.10114
    • Burn, D. J. (2002). Beyond the iron mask: towards better recognition and treatment of depression associated with Parkinson's disease. Mov. Disord. 17, 445-454. doi: 10.1002/mds.10114.
    • (2002) Mov. Disord. , vol.17 , pp. 445-454
    • Burn, D.J.1
  • 42
    • 0025794372 scopus 로고
    • Compensatory mechanisms in degenerative neurologic diseases. Insights from parkinsonism
    • doi: 10.1001/archneur.1991.00530160025009
    • Calne, D. B., and Zigmond, M. J. (1991). Compensatory mechanisms in degenerative neurologic diseases. Insights from parkinsonism. Arch. Neurol. 48, 361-363. doi: 10.1001/archneur.1991.00530160025009.
    • (1991) Arch. Neurol. , vol.48 , pp. 361-363
    • Calne, D.B.1    Zigmond, M.J.2
  • 43
    • 2442595210 scopus 로고    scopus 로고
    • Increased adenosine A2A receptors in the brain of Parkinson's disease patients with dyskinesias
    • doi: 10.1093/brain/awh128
    • Calon, F., Dridi, M., Hornykiewicz, O., Bedard, P. J., Rajput, A. H., and Di Paolo, T. (2004). Increased adenosine A2A receptors in the brain of Parkinson's disease patients with dyskinesias. Brain 127, 1075-1084. doi: 10.1093/brain/awh128.
    • (2004) Brain , vol.127 , pp. 1075-1084
    • Calon, F.1    Dridi, M.2    Hornykiewicz, O.3    Bedard, P.J.4    Rajput, A.H.5    Di Paolo, T.6
  • 44
    • 0024501249 scopus 로고
    • The NMDA antagonist MK-801 causes marked locomotor stimulation in monoamine-depleted mice
    • doi: 10.1007/BF01258633
    • Carlsson, M., and Carlsson, A. (1989). The NMDA antagonist MK-801 causes marked locomotor stimulation in monoamine-depleted mice. J. Neural. Transm. 75, 221-226. doi: 10.1007/BF01258633.
    • (1989) J. Neural. Transm. , vol.75 , pp. 221-226
    • Carlsson, M.1    Carlsson, A.2
  • 45
    • 0036828497 scopus 로고    scopus 로고
    • NMDA-receptor trafficking and targeting: implications for synaptic transmission and plasticity
    • doi: 10.1016/S0166-2236(02)02272-5
    • Carroll, R. C., and Zukin, R. S. (2002). NMDA-receptor trafficking and targeting: implications for synaptic transmission and plasticity. Trends. Neurosci. 25, 571-577. doi: 10.1016/S0166-2236(02)02272-5.
    • (2002) Trends. Neurosci. , vol.25 , pp. 571-577
    • Carroll, R.C.1    Zukin, R.S.2
  • 46
    • 81255188854 scopus 로고    scopus 로고
    • Contribution of pre-synaptic mechanisms to L-DOPA-induced dyskinesia
    • doi: 10.1016/j.neuroscience.2011.07.070
    • Carta, M., and Bezard, E. (2011). Contribution of pre-synaptic mechanisms to L-DOPA-induced dyskinesia. Neuroscience 198, 245-251. doi: 10.1016/j.neuroscience.2011.07.070.
    • (2011) Neuroscience , vol.198 , pp. 245-251
    • Carta, M.1    Bezard, E.2
  • 47
    • 34447627504 scopus 로고    scopus 로고
    • Dopamine released from 5-HT terminals is the cause of L-DOPA-induced dyskinesia in parkinsonian rats
    • doi: 10.1093/brain/awm082
    • Carta, M., Carlsson, T., Kirik, D., and Bjorklund, A. (2007). Dopamine released from 5-HT terminals is the cause of L-DOPA-induced dyskinesia in parkinsonian rats. Brain 130, 1819-1833. doi: 10.1093/brain/awm082.
    • (2007) Brain , vol.130 , pp. 1819-1833
    • Carta, M.1    Carlsson, T.2    Kirik, D.3    Bjorklund, A.4
  • 48
    • 84903895014 scopus 로고    scopus 로고
    • Ceregene Reports Data from Parkinson's Disease Phase 2b Study.
    • Ceregene, San Diego. Available online at:, [Accessed 20 July 2013].
    • Ceregene (2013). Ceregene Reports Data from Parkinson's Disease Phase 2b Study. San Diego. Available online at: http://www.ceregene.com/press_041913.asp [Accessed 20 July 2013].
    • (2013)
  • 49
    • 77953447144 scopus 로고    scopus 로고
    • A molecular basis for the increased vulnerability of substantia nigra dopamine neurons in aging and Parkinson's disease
    • doi: 10.1002/mds.22801
    • Chan, C. S., Gertler, T. S., and Surmeier, D. J. (2010). A molecular basis for the increased vulnerability of substantia nigra dopamine neurons in aging and Parkinson's disease. Mov. Disord. 25(Suppl. 1), S63-S70. doi: 10.1002/mds.22801.
    • (2010) Mov. Disord. , vol.25 , Issue.SUPPL. 1
    • Chan, C.S.1    Gertler, T.S.2    Surmeier, D.J.3
  • 50
    • 34347359673 scopus 로고    scopus 로고
    • 'Rejuvenation' protects neurons in mouse models of Parkinson's disease
    • doi: 10.1038/nature05865
    • Chan, C. S., Guzman, J. N., Ilijic, E., Mercer, J. N., Rick, C., Tkatch, T., et al. (2007). 'Rejuvenation' protects neurons in mouse models of Parkinson's disease. Nature 447, 1081-1086. doi: 10.1038/nature05865.
    • (2007) Nature , vol.447 , pp. 1081-1086
    • Chan, C.S.1    Guzman, J.N.2    Ilijic, E.3    Mercer, J.N.4    Rick, C.5    Tkatch, T.6
  • 51
    • 0035874345 scopus 로고    scopus 로고
    • Neuroprotection by caffeine and A(2A) adenosine receptor inactivation in a model of Parkinson's disease
    • Chen, J. F., Xu, K., Petzer, J. P., Staal, R., Xu, Y. H., Beilstein, M., et al. (2001). Neuroprotection by caffeine and A(2A) adenosine receptor inactivation in a model of Parkinson's disease. J. Neurosci. 21, RC143..
    • (2001) J. Neurosci. , vol.21
    • Chen, J.F.1    Xu, K.2    Petzer, J.P.3    Staal, R.4    Xu, Y.H.5    Beilstein, M.6
  • 52
    • 0024093449 scopus 로고
    • Glutamate neurotoxicity and diseases of the nervous system
    • doi: 10.1016/0896-6273(88)90162-6
    • Choi, D. W. (1988). Glutamate neurotoxicity and diseases of the nervous system. Neuron 1, 623-634. doi: 10.1016/0896-6273(88)90162-6.
    • (1988) Neuron , vol.1 , pp. 623-634
    • Choi, D.W.1
  • 53
    • 73449128979 scopus 로고    scopus 로고
    • Safety and tolerability of putaminal AADC gene therapy for Parkinson disease
    • doi: 10.1212/WNL.0b013e3181c29356
    • Christine, C. W., Starr, P. A., Larson, P. S., Eberling, J. L., Jagust, W. J., Hawkins, R. A., et al. (2009). Safety and tolerability of putaminal AADC gene therapy for Parkinson disease. Neurology 73, 1662-1669. doi: 10.1212/WNL.0b013e3181c29356.
    • (2009) Neurology , vol.73 , pp. 1662-1669
    • Christine, C.W.1    Starr, P.A.2    Larson, P.S.3    Eberling, J.L.4    Jagust, W.J.5    Hawkins, R.A.6
  • 54
    • 26944447877 scopus 로고    scopus 로고
    • Systemic exposure to paraquat and maneb models early Parkinson's disease in young adult rats
    • doi: 10.1016/j.nbd.2005.03.018
    • Cicchetti, F., Lapointe, N., Roberge-Tremblay, A., Saint-Pierre, M., Jimenez, L., Ficke, B. W., et al. (2005). Systemic exposure to paraquat and maneb models early Parkinson's disease in young adult rats. Neurobiol. Dis. 20, 360-371. doi: 10.1016/j.nbd.2005.03.018.
    • (2005) Neurobiol. Dis. , vol.20 , pp. 360-371
    • Cicchetti, F.1    Lapointe, N.2    Roberge-Tremblay, A.3    Saint-Pierre, M.4    Jimenez, L.5    Ficke, B.W.6
  • 55
    • 84867051523 scopus 로고    scopus 로고
    • Tumor necrosis factor-induced cerebral insulin resistance in Alzheimer's disease links numerous treatment rationales
    • doi: 10.1124/pr.112.005850
    • Clark, I., Atwood, C., Bowen, R., Paz-Filho, G., and Vissel, B. (2012). Tumor necrosis factor-induced cerebral insulin resistance in Alzheimer's disease links numerous treatment rationales. Pharmacol. Rev. 64, 1004-1026. doi: 10.1124/pr.112.005850.
    • (2012) Pharmacol. Rev. , vol.64 , pp. 1004-1026
    • Clark, I.1    Atwood, C.2    Bowen, R.3    Paz-Filho, G.4    Vissel, B.5
  • 56
    • 78049266766 scopus 로고    scopus 로고
    • The roles of TNF in brain dysfunction and disease
    • doi: 10.1016/j.pharmthera.2010.08.007
    • Clark, I. A., Alleva, L. M., and Vissel, B. (2010). The roles of TNF in brain dysfunction and disease. Pharmacol. Ther. 128, 519-548. doi: 10.1016/j.pharmthera.2010.08.007.
    • (2010) Pharmacol. Ther. , vol.128 , pp. 519-548
    • Clark, I.A.1    Alleva, L.M.2    Vissel, B.3
  • 57
    • 84884704633 scopus 로고    scopus 로고
    • Treatment implications of the altered cytokine-insulin axis in neurodegenerative disease
    • doi: 10.1016/j.bcp.2013.07.030
    • Clark, I. A., and Vissel, B. (2013). Treatment implications of the altered cytokine-insulin axis in neurodegenerative disease. Biochem. Pharmacol. 86, 862-871. doi: 10.1016/j.bcp.2013.07.030.
    • (2013) Biochem. Pharmacol. , vol.86 , pp. 862-871
    • Clark, I.A.1    Vissel, B.2
  • 58
    • 3242765264 scopus 로고    scopus 로고
    • Simultaneous blockade of adenosine A2A and metabotropic glutamate mGlu5 receptors increase their efficacy in reversing Parkinsonian deficits in rats
    • doi: 10.1038/sj.npp.1300444
    • Coccurello, R., Breysse, N., and Amalric, M. (2004). Simultaneous blockade of adenosine A2A and metabotropic glutamate mGlu5 receptors increase their efficacy in reversing Parkinsonian deficits in rats. Neuropsychopharmacology 29, 1451-1461. doi: 10.1038/sj.npp.1300444.
    • (2004) Neuropsychopharmacology , vol.29 , pp. 1451-1461
    • Coccurello, R.1    Breysse, N.2    Amalric, M.3
  • 59
    • 0032936111 scopus 로고    scopus 로고
    • Therapeutic potential of nerve growth factors in Parkinson's disease
    • doi: 10.2165/00002512-199914040-00003
    • Collier, T. J., and Sortwell, C. E. (1999). Therapeutic potential of nerve growth factors in Parkinson's disease. Drugs Aging. 14, 261-287. doi: 10.2165/00002512-199914040-00003.
    • (1999) Drugs Aging. , vol.14 , pp. 261-287
    • Collier, T.J.1    Sortwell, C.E.2
  • 60
    • 27944470972 scopus 로고    scopus 로고
    • Metabotropic glutamate receptors in the basal ganglia motor circuit
    • doi: 10.1038/nrn1763
    • Conn, P. J., Battaglia, G., Marino, M. J., and Nicoletti, F. (2005). Metabotropic glutamate receptors in the basal ganglia motor circuit. Nat. Rev. Neurosci. 6, 787-798. doi: 10.1038/nrn1763.
    • (2005) Nat. Rev. Neurosci. , vol.6 , pp. 787-798
    • Conn, P.J.1    Battaglia, G.2    Marino, M.J.3    Nicoletti, F.4
  • 61
    • 0031838816 scopus 로고    scopus 로고
    • Paraquat: a useful tool for the in vivo study of mechanisms of neuronal cell death
    • doi: 10.1111/j.1600-0773.1998.tb01434.x
    • Corasaniti, M. T., Strongoli, M. C., Rotiroti, D., Bagetta, G., and Nistico, G. (1998). Paraquat: a useful tool for the in vivo study of mechanisms of neuronal cell death. Pharmacol. Toxicol. 83, 1-7. doi: 10.1111/j.1600-0773.1998.tb01434.x.
    • (1998) Pharmacol. Toxicol. , vol.83 , pp. 1-7
    • Corasaniti, M.T.1    Strongoli, M.C.2    Rotiroti, D.3    Bagetta, G.4    Nistico, G.5
  • 62
    • 70449519036 scopus 로고    scopus 로고
    • Oxidative stress and cell death in cells expressing L-ferritin variants causing neuroferritinopathy
    • doi: 10.1016/j.nbd.2009.09.009
    • Cozzi, A., Rovelli, E., Frizzale, G., Campanella, A., Amendola, M., Arosio, P., et al. (2010). Oxidative stress and cell death in cells expressing L-ferritin variants causing neuroferritinopathy. Neurobiol. Dis. 37, 77-85. doi: 10.1016/j.nbd.2009.09.009.
    • (2010) Neurobiol. Dis. , vol.37 , pp. 77-85
    • Cozzi, A.1    Rovelli, E.2    Frizzale, G.3    Campanella, A.4    Amendola, M.5    Arosio, P.6
  • 63
    • 84895904524 scopus 로고    scopus 로고
    • Pimavanserin for patients with Parkinson's disease psychosis: a randomised, placebo-controlled phase 3 trial
    • doi: 10.1016/S0140-6736(13)62106-6
    • Cummings, J., Isaacson, S., Mills, R., Williams, H., Chi-Burris, K., Corbett, A., et al. (2013). Pimavanserin for patients with Parkinson's disease psychosis: a randomised, placebo-controlled phase 3 trial. Lancet 383, 533-540. doi: 10.1016/S0140-6736(13)62106-6.
    • (2013) Lancet , vol.383 , pp. 533-540
    • Cummings, J.1    Isaacson, S.2    Mills, R.3    Williams, H.4    Chi-Burris, K.5    Corbett, A.6
  • 64
    • 0141741347 scopus 로고    scopus 로고
    • Parkinson's disease: mechanisms and models
    • doi: 10.1016/S0896-6273(03)00568-3
    • Dauer, W., and Przedborski, S. (2003). Parkinson's disease: mechanisms and models. Neuron 39, 889-909. doi: 10.1016/S0896-6273(03)00568-3.
    • (2003) Neuron , vol.39 , pp. 889-909
    • Dauer, W.1    Przedborski, S.2
  • 65
    • 33645050113 scopus 로고    scopus 로고
    • Effects of group I metabotropic glutamate receptors blockade in experimental models of Parkinson's disease
    • doi: 10.1016/j.brainresbull.2005.12.009
    • Dekundy, A., Pietraszek, M., Schaefer, D., Cenci, M. A., and Danysz, W. (2006). Effects of group I metabotropic glutamate receptors blockade in experimental models of Parkinson's disease. Brain Res. Bull. 69, 318-326. doi: 10.1016/j.brainresbull.2005.12.009.
    • (2006) Brain Res. Bull. , vol.69 , pp. 318-326
    • Dekundy, A.1    Pietraszek, M.2    Schaefer, D.3    Cenci, M.A.4    Danysz, W.5
  • 66
    • 84869830250 scopus 로고    scopus 로고
    • Leucine-rich repeat kinase 2 inhibitors: a patent review (2006 - 2011)
    • doi: 10.1517/13543776.2012.729041
    • Deng, X., Choi, H. G., Buhrlage, S. J., and Gray, N. S. (2012). Leucine-rich repeat kinase 2 inhibitors: a patent review (2006 - 2011). Expert Opin. Ther. Pat. 22, 1415-1426. doi: 10.1517/13543776.2012.729041.
    • (2012) Expert Opin. Ther. Pat. , vol.22 , pp. 1415-1426
    • Deng, X.1    Choi, H.G.2    Buhrlage, S.J.3    Gray, N.S.4
  • 67
    • 33846558995 scopus 로고    scopus 로고
    • Regulatory mechanisms of AMPA receptors in synaptic plasticity
    • doi: 10.1038/nrn2055
    • Derkach, V. A., Oh, M. C., Guire, E. S., and Soderling, T. R. (2007). Regulatory mechanisms of AMPA receptors in synaptic plasticity. Nat. Rev. Neurosci. 8, 101-113. doi: 10.1038/nrn2055.
    • (2007) Nat. Rev. Neurosci. , vol.8 , pp. 101-113
    • Derkach, V.A.1    Oh, M.C.2    Guire, E.S.3    Soderling, T.R.4
  • 68
    • 84897930725 scopus 로고    scopus 로고
    • Targeting chelatable iron as a therapeutic modality in Parkinson's disease.
    • doi: 10.1089/ars.2013.5593. [Epub ahead of print].
    • Devos, D., Moreau, C., Devedjian, J. C., Kluza, J., Petrault, M., Laloux, C., et al. (2014). Targeting chelatable iron as a therapeutic modality in Parkinson's disease. Antioxid. Redox. Signal. doi: 10.1089/ars.2013.5593. [Epub ahead of print]..
    • (2014) Antioxid. Redox. Signal.
    • Devos, D.1    Moreau, C.2    Devedjian, J.C.3    Kluza, J.4    Petrault, M.5    Laloux, C.6
  • 70
    • 49049112072 scopus 로고    scopus 로고
    • Evidence for segregated and integrative connectivity patterns in the human Basal Ganglia
    • doi: 10.1523/JNEUROSCI.1486-08.2008
    • Draganski, B., Kherif, F., Kloppel, S., Cook, P. A., Alexander, D. C., Parker, G. J., et al. (2008). Evidence for segregated and integrative connectivity patterns in the human Basal Ganglia. J. Neurosci. 28, 7143-7152. doi: 10.1523/JNEUROSCI.1486-08.2008.
    • (2008) J. Neurosci. , vol.28 , pp. 7143-7152
    • Draganski, B.1    Kherif, F.2    Kloppel, S.3    Cook, P.A.4    Alexander, D.C.5    Parker, G.J.6
  • 71
    • 1042299831 scopus 로고    scopus 로고
    • Clozapine improves dyskinesias in Parkinson disease: a double-blind, placebo-controlled study
    • doi: 10.1212/01.WNL.0000110317.52453.6C
    • Durif, F., Debilly, B., Galitzky, M., Morand, D., Viallet, F., Borg, M., et al. (2004). Clozapine improves dyskinesias in Parkinson disease: a double-blind, placebo-controlled study. Neurology 62, 381-388. doi: 10.1212/01.WNL.0000110317.52453.6C.
    • (2004) Neurology , vol.62 , pp. 381-388
    • Durif, F.1    Debilly, B.2    Galitzky, M.3    Morand, D.4    Viallet, F.5    Borg, M.6
  • 72
    • 0030941117 scopus 로고    scopus 로고
    • Low-dose clozapine improves dyskinesias in Parkinson's disease
    • doi: 10.1212/WNL.48.3.658
    • Durif, F., Vidailhet, M., Assal, F., Roche, C., Bonnet, A. M., and Agid, Y. (1997). Low-dose clozapine improves dyskinesias in Parkinson's disease. Neurology 48, 658-662. doi: 10.1212/WNL.48.3.658.
    • (1997) Neurology , vol.48 , pp. 658-662
    • Durif, F.1    Vidailhet, M.2    Assal, F.3    Roche, C.4    Bonnet, A.M.5    Agid, Y.6
  • 74
    • 80052533576 scopus 로고    scopus 로고
    • Animal models of Parkinson's disease: a source of novel treatments and clues to the cause of the disease
    • doi: 10.1111/j.1476-5381.2011.01426.x
    • Duty, S., and Jenner, P. (2011). Animal models of Parkinson's disease: a source of novel treatments and clues to the cause of the disease. Br. J. Pharmacol. 164, 1357-1391. doi: 10.1111/j.1476-5381.2011.01426.x.
    • (2011) Br. J. Pharmacol. , vol.164 , pp. 1357-1391
    • Duty, S.1    Jenner, P.2
  • 75
    • 66149096521 scopus 로고    scopus 로고
    • Functional effects of AAV2-GDNF on the dopaminergic nigrostriatal pathway in parkinsonian rhesus monkeys
    • doi: 10.1089/hum.2008.201
    • Eberling, J. L., Kells, A. P., Pivirotto, P., Beyer, J., Bringas, J., Federoff, H. J., et al. (2009). Functional effects of AAV2-GDNF on the dopaminergic nigrostriatal pathway in parkinsonian rhesus monkeys. Hum. Gene. Ther. 20, 511-518. doi: 10.1089/hum.2008.201.
    • (2009) Hum. Gene. Ther. , vol.20 , pp. 511-518
    • Eberling, J.L.1    Kells, A.P.2    Pivirotto, P.3    Beyer, J.4    Bringas, J.5    Federoff, H.J.6
  • 76
    • 77951958360 scopus 로고    scopus 로고
    • Safety and efficacy of perampanel in advanced Parkinson's disease: a randomized, placebo-controlled study
    • doi: 10.1002/mds.22974
    • Eggert, K., Squillacote, D., Barone, P., Dodel, R., Katzenschlager, R., Emre, M., et al. (2010). Safety and efficacy of perampanel in advanced Parkinson's disease: a randomized, placebo-controlled study. Mov. Disord. 25, 896-905. doi: 10.1002/mds.22974.
    • (2010) Mov. Disord. , vol.25 , pp. 896-905
    • Eggert, K.1    Squillacote, D.2    Barone, P.3    Dodel, R.4    Katzenschlager, R.5    Emre, M.6
  • 77
    • 0034023862 scopus 로고    scopus 로고
    • The stimulant effects of caffeine on locomotor behaviour in mice are mediated through its blockade of adenosine A(2A) receptors
    • doi: 10.1038/sj.bjp.0703170
    • El Yacoubi, M., Ledent, C., Menard, J. F., Parmentier, M., Costentin, J., and Vaugeois, J. M. (2000). The stimulant effects of caffeine on locomotor behaviour in mice are mediated through its blockade of adenosine A(2A) receptors. Br. J. Pharmacol. 129, 1465-1473. doi: 10.1038/sj.bjp.0703170.
    • (2000) Br. J. Pharmacol. , vol.129 , pp. 1465-1473
    • El Yacoubi, M.1    Ledent, C.2    Menard, J.F.3    Parmentier, M.4    Costentin, J.5    Vaugeois, J.M.6
  • 78
    • 33847198228 scopus 로고    scopus 로고
    • Subthalamic glutamic acid decarboxylase gene therapy: changes in motor function and cortical metabolism
    • doi: 10.1038/sj.jcbfm.9600364
    • Emborg, M. E., Carbon, M., Holden, J. E., During, M. J., Ma, Y., Tang, C., et al. (2007). Subthalamic glutamic acid decarboxylase gene therapy: changes in motor function and cortical metabolism. J. Cereb. Blood. Flow. Metab. 27, 501-509. doi: 10.1038/sj.jcbfm.9600364.
    • (2007) J. Cereb. Blood. Flow. Metab. , vol.27 , pp. 501-509
    • Emborg, M.E.1    Carbon, M.2    Holden, J.E.3    During, M.J.4    Ma, Y.5    Tang, C.6
  • 79
    • 0042970162 scopus 로고    scopus 로고
    • Dementia associated with Parkinson's disease
    • doi: 10.1016/S1474-4422(03)00351-X
    • Emre, M. (2003). Dementia associated with Parkinson's disease. Lancet Neurol. 2, 229-237. doi: 10.1016/S1474-4422(03)00351-X.
    • (2003) Lancet Neurol. , vol.2 , pp. 229-237
    • Emre, M.1
  • 80
    • 77956874908 scopus 로고    scopus 로고
    • Memantine for patients with Parkinson's disease dementia or dementia with Lewy bodies: a randomised, double-blind, placebo-controlled trial
    • doi: 10.1016/S1474-4422(10)70194-0
    • Emre, M., Tsolaki, M., Bonuccelli, U., Destee, A., Tolosa, E., Kutzelnigg, A., et al. (2010). Memantine for patients with Parkinson's disease dementia or dementia with Lewy bodies: a randomised, double-blind, placebo-controlled trial. Lancet Neurol. 9, 969-977. doi: 10.1016/S1474-4422(10)70194-0.
    • (2010) Lancet Neurol. , vol.9 , pp. 969-977
    • Emre, M.1    Tsolaki, M.2    Bonuccelli, U.3    Destee, A.4    Tolosa, E.5    Kutzelnigg, A.6
  • 81
    • 0028556522 scopus 로고
    • NMDA receptor blockade reverses motor response alterations induced by levodopa
    • doi: 10.1097/00001756-199412000-00045
    • Engber, T. M., Papa, S. M., Boldry, R. C., and Chase, T. N. (1994). NMDA receptor blockade reverses motor response alterations induced by levodopa. Neuroreport 5, 2586-2588. doi: 10.1097/00001756-199412000-00045.
    • (1994) Neuroreport , vol.5 , pp. 2586-2588
    • Engber, T.M.1    Papa, S.M.2    Boldry, R.C.3    Chase, T.N.4
  • 82
    • 0022445894 scopus 로고
    • Effects of adult or neonatal treatment with 6-hydroxydopamine or 5,7-dihydroxytryptamine on locomotor activity, monoamine levels, and response to caffeine
    • doi: 10.1016/0091-3057(86)90453-3
    • Erinoff, L., and Snodgrass, S. R. (1986). Effects of adult or neonatal treatment with 6-hydroxydopamine or 5,7-dihydroxytryptamine on locomotor activity, monoamine levels, and response to caffeine. Pharmacol. Biochem. Behav. 24, 1039-1045. doi: 10.1016/0091-3057(86)90453-3.
    • (1986) Pharmacol. Biochem. Behav. , vol.24 , pp. 1039-1045
    • Erinoff, L.1    Snodgrass, S.R.2
  • 83
    • 0345257314 scopus 로고    scopus 로고
    • Recombinant adeno-associated viral vector (rAAV) delivery of GDNF provides protection against 6-OHDA lesion in the common marmoset monkey (Callithrix jacchus)
    • doi: 10.1016/j.expneurol.2003.08.007
    • Eslamboli, A., Cummings, R. M., Ridley, R. M., Baker, H. F., Muzyczka, N., Burger, C., et al. (2003). Recombinant adeno-associated viral vector (rAAV) delivery of GDNF provides protection against 6-OHDA lesion in the common marmoset monkey (Callithrix jacchus). Exp. Neurol. 184, 536-548. doi: 10.1016/j.expneurol.2003.08.007.
    • (2003) Exp. Neurol. , vol.184 , pp. 536-548
    • Eslamboli, A.1    Cummings, R.M.2    Ridley, R.M.3    Baker, H.F.4    Muzyczka, N.5    Burger, C.6
  • 84
    • 77953618656 scopus 로고    scopus 로고
    • A long-term study of istradefylline in subjects with fluctuating Parkinson's disease
    • doi: 10.1016/j.parkreldis.2010.02.014
    • Factor, S., Mark, M. H., Watts, R., Struck, L., Mori, A., Ballerini, R., et al. (2010). A long-term study of istradefylline in subjects with fluctuating Parkinson's disease. Parkinsonism. Relat. Disord. 16, 423-426. doi: 10.1016/j.parkreldis.2010.02.014.
    • (2010) Parkinsonism. Relat. Disord. , vol.16 , pp. 423-426
    • Factor, S.1    Mark, M.H.2    Watts, R.3    Struck, L.4    Mori, A.5    Ballerini, R.6
  • 85
    • 84879604899 scopus 로고    scopus 로고
    • Long-term safety and efficacy of preladenant in subjects with fluctuating Parkinson's disease
    • doi: 10.1002/mds.25395
    • Factor, S. A., Wolski, K., Togasaki, D. M., Huyck, S., Cantillon, M., Ho, T. W., et al. (2013). Long-term safety and efficacy of preladenant in subjects with fluctuating Parkinson's disease. Mov. Disord. 28, 817-820. doi: 10.1002/mds.25395.
    • (2013) Mov. Disord. , vol.28 , pp. 817-820
    • Factor, S.A.1    Wolski, K.2    Togasaki, D.M.3    Huyck, S.4    Cantillon, M.5    Ho, T.W.6
  • 86
    • 0032553713 scopus 로고    scopus 로고
    • Behavioral recovery in 6-hydroxydopamine-lesioned rats by cotransduction of striatum with tyrosine hydroxylase and aromatic L-amino acid decarboxylase genes using two separate adeno-associated virus vectors
    • doi: 10.1089/hum.1998.9.17-2527
    • Fan, D. S., Ogawa, M., Fujimoto, K. I., Ikeguchi, K., Ogasawara, Y., Urabe, M., et al. (1998). Behavioral recovery in 6-hydroxydopamine-lesioned rats by cotransduction of striatum with tyrosine hydroxylase and aromatic L-amino acid decarboxylase genes using two separate adeno-associated virus vectors. Hum. Gene. Ther. 9, 2527-2535. doi: 10.1089/hum.1998.9.17-2527.
    • (1998) Hum. Gene. Ther. , vol.9 , pp. 2527-2535
    • Fan, D.S.1    Ogawa, M.2    Fujimoto, K.I.3    Ikeguchi, K.4    Ogasawara, Y.5    Urabe, M.6
  • 87
    • 0032862588 scopus 로고    scopus 로고
    • Salicylate protects against MPTP-induced impairments in dopaminergic neurotransmission at the striatal and nigral level in mice
    • doi: 10.1007/s002109900079
    • Ferger, B., Teismann, P., Earl, C. D., Kuschinsky, K., and Oertel, W. H. (1999). Salicylate protects against MPTP-induced impairments in dopaminergic neurotransmission at the striatal and nigral level in mice. Naunyn. Schmiedebergs. Arch. Pharmacol. 360, 256-261. doi: 10.1007/s002109900079.
    • (1999) Naunyn. Schmiedebergs. Arch. Pharmacol. , vol.360 , pp. 256-261
    • Ferger, B.1    Teismann, P.2    Earl, C.D.3    Kuschinsky, K.4    Oertel, W.H.5
  • 88
    • 26844527655 scopus 로고    scopus 로고
    • Genetic mouse models of parkinsonism: strengths and limitations
    • doi: 10.1602/neurorx.2.3.495
    • Fleming, S. M., Fernagut, P. O., and Chesselet, M. F. (2005). Genetic mouse models of parkinsonism: strengths and limitations. NeuroRx 2, 495-503. doi: 10.1602/neurorx.2.3.495.
    • (2005) NeuroRx , vol.2 , pp. 495-503
    • Fleming, S.M.1    Fernagut, P.O.2    Chesselet, M.F.3
  • 89
    • 84904963738 scopus 로고    scopus 로고
    • Azilect (Rasagiline Mesylate).
    • Food Drug Administration, Silver Spring, MD
    • Food and Drug Administration (2011). Azilect (Rasagiline Mesylate). Silver Spring, MD.
    • (2011)
  • 90
    • 35448979969 scopus 로고    scopus 로고
    • Noradrenaline in Parkinson's disease: from disease progression to current therapeutics
    • doi: 10.2174/092986707781745550
    • Fornai, F., Di Poggio, A. B., Pellegrini, A., Ruggieri, S., and Paparelli, A. (2007). Noradrenaline in Parkinson's disease: from disease progression to current therapeutics. Curr. Med. Chem. 14, 2330-2334. doi: 10.2174/092986707781745550.
    • (2007) Curr. Med. Chem. , vol.14 , pp. 2330-2334
    • Fornai, F.1    Di Poggio, A.B.2    Pellegrini, A.3    Ruggieri, S.4    Paparelli, A.5
  • 91
    • 0035412923 scopus 로고    scopus 로고
    • Neural mechanisms underlying peak-dose dyskinesia induced by levodopa and apomorphine are distinct: evidence from the effects of the alpha(2) adrenoceptor antagonist idazoxan
    • doi: 10.1002/mds.1148
    • Fox, S. H., Henry, B., Hill, M. P., Peggs, D., Crossman, A. R., and Brotchie, J. M. (2001). Neural mechanisms underlying peak-dose dyskinesia induced by levodopa and apomorphine are distinct: evidence from the effects of the alpha(2) adrenoceptor antagonist idazoxan. Mov. Disord. 16, 642-650. doi: 10.1002/mds.1148.
    • (2001) Mov. Disord. , vol.16 , pp. 642-650
    • Fox, S.H.1    Henry, B.2    Hill, M.P.3    Peggs, D.4    Crossman, A.R.5    Brotchie, J.M.6
  • 92
    • 77949899236 scopus 로고    scopus 로고
    • Anti-inflammatory drugs and risk of Parkinson disease: a meta-analysis
    • doi: 10.1212/WNL.0b013e3181d5a4a3
    • Gagne, J. J., and Power, M. C. (2010). Anti-inflammatory drugs and risk of Parkinson disease: a meta-analysis. Neurology 74, 995-1002. doi: 10.1212/WNL.0b013e3181d5a4a3.
    • (2010) Neurology , vol.74 , pp. 995-1002
    • Gagne, J.J.1    Power, M.C.2
  • 93
    • 25644452043 scopus 로고    scopus 로고
    • Novel multifunctional neuroprotective iron chelator-monoamine oxidase inhibitor drugs for neurodegenerative diseases. In vivo selective brain monoamine oxidase inhibition and prevention of MPTP-induced striatal dopamine depletion.
    • doi: 10.1111/j.1471-4159.2005.03341.x
    • Gal, S., Zheng, H., Fridkin, M., and Youdim, M. B. (2005). Novel multifunctional neuroprotective iron chelator-monoamine oxidase inhibitor drugs for neurodegenerative diseases. In vivo selective brain monoamine oxidase inhibition and prevention of MPTP-induced striatal dopamine depletion. J. Neurochem. 95, 79-88. doi: 10.1111/j.1471-4159.2005.03341.x.
    • (2005) J. Neurochem. , vol.95 , pp. 79-88
    • Gal, S.1    Zheng, H.2    Fridkin, M.3    Youdim, M.B.4
  • 94
    • 47749126515 scopus 로고    scopus 로고
    • Why neurodegenerative diseases are progressive: uncontrolled inflammation drives disease progression
    • doi: 10.1016/j.it.2008.05.002
    • Gao, H. M., and Hong, J. S. (2008). Why neurodegenerative diseases are progressive: uncontrolled inflammation drives disease progression. Trends. Immunol. 29, 357-365. doi: 10.1016/j.it.2008.05.002.
    • (2008) Trends. Immunol. , vol.29 , pp. 357-365
    • Gao, H.M.1    Hong, J.S.2
  • 95
    • 0037375896 scopus 로고    scopus 로고
    • Parkinson's disease and sleep
    • doi: 10.1053/smrv.2002.0229
    • Garcia-Borreguero, D., Larrosa, O., and Bravo, M. (2003). Parkinson's disease and sleep. Sleep. Med. Rev. 7, 115-129. doi: 10.1053/smrv.2002.0229.
    • (2003) Sleep. Med. Rev. , vol.7 , pp. 115-129
    • Garcia-Borreguero, D.1    Larrosa, O.2    Bravo, M.3
  • 96
    • 34250336916 scopus 로고    scopus 로고
    • AAV2-mediated delivery of human neurturin to the rat nigrostriatal system: long-term efficacy tolerability of CERE-120 for Parkinson's disease.
    • doi: 10.1016/j.nbd.2007.04.003
    • Gasmi, M., Brandon, E. P., Herzog, C. D., Wilson, A., Bishop, K. M., Hofer, E. K., et al. (2007a). AAV2-mediated delivery of human neurturin to the rat nigrostriatal system: long-term efficacy and tolerability of CERE-120 for Parkinson's disease. Neurobiol. Dis. 27, 67-76. doi: 10.1016/j.nbd.2007.04.003.
    • (2007) Neurobiol. Dis. , vol.27 , pp. 67-76
    • Gasmi, M.1    Brandon, E.P.2    Herzog, C.D.3    Wilson, A.4    Bishop, K.M.5    Hofer, E.K.6
  • 97
    • 33846011117 scopus 로고    scopus 로고
    • Striatal delivery of neurturin by CERE-120, an AAV2 vector for the treatment of dopaminergic neuron degeneration in Parkinson's disease.
    • doi: 10.1038/sj.mt.6300010
    • Gasmi, M., Herzog, C. D., Brandon, E. P., Cunningham, J. J., Ramirez, G. A., Ketchum, E. T., et al. (2007b). Striatal delivery of neurturin by CERE-120, an AAV2 vector for the treatment of dopaminergic neuron degeneration in Parkinson's disease. Mol. Ther. 15, 62-68. doi: 10.1038/sj.mt.6300010.
    • (2007) Mol. Ther. , vol.15 , pp. 62-68
    • Gasmi, M.1    Herzog, C.D.2    Brandon, E.P.3    Cunningham, J.J.4    Ramirez, G.A.5    Ketchum, E.T.6
  • 98
    • 0026531301 scopus 로고
    • The neostriatal mosaic: multiple levels of compartmental organization
    • doi: 10.1016/0166-2236(92)90355-C
    • Gerfen, C. R. (1992). The neostriatal mosaic: multiple levels of compartmental organization. Trends. Neurosci. 15, 133-139. doi: 10.1016/0166-2236(92)90355-C.
    • (1992) Trends. Neurosci. , vol.15 , pp. 133-139
    • Gerfen, C.R.1
  • 99
    • 0033616388 scopus 로고    scopus 로고
    • GDNF improves dopamine function in the substantia nigra but not the putamen of unilateral MPTP-lesioned rhesus monkeys
    • doi: 10.1016/S0006-8993(98)01244-X
    • Gerhardt, G. A., Cass, W. A., Huettl, P., Brock, S., Zhang, Z., and Gash, D. M. (1999). GDNF improves dopamine function in the substantia nigra but not the putamen of unilateral MPTP-lesioned rhesus monkeys. Brain Res. 817, 163-171. doi: 10.1016/S0006-8993(98)01244-X.
    • (1999) Brain Res. , vol.817 , pp. 163-171
    • Gerhardt, G.A.1    Cass, W.A.2    Huettl, P.3    Brock, S.4    Zhang, Z.5    Gash, D.M.6
  • 100
    • 33847757879 scopus 로고    scopus 로고
    • Sarizotan as a treatment for dyskinesias in Parkinson's disease: a double-blind placebo-controlled trial
    • doi: 10.1002/mds.21226
    • Goetz, C. G., Damier, P., Hicking, C., Laska, E., Muller, T., Olanow, C. W., et al. (2007). Sarizotan as a treatment for dyskinesias in Parkinson's disease: a double-blind placebo-controlled trial. Mov. Disord. 22, 179-186. doi: 10.1002/mds.21226.
    • (2007) Mov. Disord. , vol.22 , pp. 179-186
    • Goetz, C.G.1    Damier, P.2    Hicking, C.3    Laska, E.4    Muller, T.5    Olanow, C.W.6
  • 101
    • 0027384928 scopus 로고
    • Effect of nondopaminergic drugs on L-dopa-induced dyskinesias in MPTP-treated monkeys
    • doi: 10.1097/00002826-199310000-00004
    • Gomez-Mancilla, B., and Bedard, P. J. (1993). Effect of nondopaminergic drugs on L-dopa-induced dyskinesias in MPTP-treated monkeys. Clin. Neuropharmacol. 16, 418-427. doi: 10.1097/00002826-199310000-00004.
    • (1993) Clin. Neuropharmacol. , vol.16 , pp. 418-427
    • Gomez-Mancilla, B.1    Bedard, P.J.2
  • 102
    • 17144446935 scopus 로고
    • Injection of excitatory amino acid antagonists into the medial pallidal segment of a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treated primate reverses motor symptoms of parkinsonism
    • doi: 10.1016/0024-3205(90)90376-3
    • Graham, W. C., Robertson, R. G., Sambrook, M. A., and Crossman, A. R. (1990). Injection of excitatory amino acid antagonists into the medial pallidal segment of a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treated primate reverses motor symptoms of parkinsonism. Life Sci. 47, PL91-PL97. doi: 10.1016/0024-3205(90)90376-3.
    • (1990) Life Sci. , vol.47
    • Graham, W.C.1    Robertson, R.G.2    Sambrook, M.A.3    Crossman, A.R.4
  • 103
    • 0025293685 scopus 로고
    • Neurotransmitters and neuromodulators in the basal ganglia
    • doi: 10.1016/0166-2236(90)90104-I
    • Graybiel, A. M. (1990). Neurotransmitters and neuromodulators in the basal ganglia. Trends Neurosci. 13, 244-254. doi: 10.1016/0166-2236(90)90104-I.
    • (1990) Trends Neurosci. , vol.13 , pp. 244-254
    • Graybiel, A.M.1
  • 104
    • 0034941949 scopus 로고    scopus 로고
    • Gene expression analysis in N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mice model of Parkinson's disease using cDNA microarray: effect of R-apomorphine
    • doi: 10.1046/j.1471-4159.2001.00397.x
    • Grunblatt, E., Mandel, S., Maor, G., and Youdim, M. B. (2001). Gene expression analysis in N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mice model of Parkinson's disease using cDNA microarray: effect of R-apomorphine. J. Neurochem. 78, 1-12. doi: 10.1046/j.1471-4159.2001.00397.x.
    • (2001) J. Neurochem. , vol.78 , pp. 1-12
    • Grunblatt, E.1    Mandel, S.2    Maor, G.3    Youdim, M.B.4
  • 105
    • 0043127452 scopus 로고    scopus 로고
    • Selective antagonism of GluR5 kainate-receptor-mediated synaptic currents by topiramate in rat basolateral amygdala neurons
    • Gryder, D. S., and Rogawski, M. A. (2003). Selective antagonism of GluR5 kainate-receptor-mediated synaptic currents by topiramate in rat basolateral amygdala neurons. J. Neurosci. 23, 7069-7074..
    • (2003) J. Neurosci. , vol.23 , pp. 7069-7074
    • Gryder, D.S.1    Rogawski, M.A.2
  • 106
    • 0023244667 scopus 로고
    • Reversible serotoninergic neurotoxicity of N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in mouse striatum studied by neurochemical and immunohistochemical approaches
    • doi: 10.1016/0006-8993(87)90341-6
    • Hara, K., Tohyama, I., Kimura, H., Fukuda, H., Nakamura, S., and Kameyama, M. (1987). Reversible serotoninergic neurotoxicity of N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in mouse striatum studied by neurochemical and immunohistochemical approaches. Brain Res. 410, 371-374. doi: 10.1016/0006-8993(87)90341-6.
    • (1987) Brain Res. , vol.410 , pp. 371-374
    • Hara, K.1    Tohyama, I.2    Kimura, H.3    Fukuda, H.4    Nakamura, S.5    Kameyama, M.6
  • 107
    • 77956917231 scopus 로고    scopus 로고
    • Synaptic versus extrasynaptic NMDA receptor signalling: implications for neurodegenerative disorders
    • doi: 10.1038/nrn2911
    • Hardingham, G. E., and Bading, H. (2010). Synaptic versus extrasynaptic NMDA receptor signalling: implications for neurodegenerative disorders. Nat. Rev. Neurosci. 11, 682-696. doi: 10.1038/nrn2911.
    • (2010) Nat. Rev. Neurosci. , vol.11 , pp. 682-696
    • Hardingham, G.E.1    Bading, H.2
  • 108
    • 45549108268 scopus 로고    scopus 로고
    • Glucagon-like peptide 1 receptor stimulation reverses key deficits in distinct rodent models of Parkinson's disease.
    • doi: 10.1186/1742-2094-5-19
    • Harkavyi, A., Abuirmeileh, A., Lever, R., Kingsbury, A. E., Biggs, C. S., and Whitton, P. S. (2008). Glucagon-like peptide 1 receptor stimulation reverses key deficits in distinct rodent models of Parkinson's disease. J. Neuroinflammation 5:19. doi: 10.1186/1742-2094-5-19.
    • (2008) J. Neuroinflammation , vol.5 , pp. 19
    • Harkavyi, A.1    Abuirmeileh, A.2    Lever, R.3    Kingsbury, A.E.4    Biggs, C.S.5    Whitton, P.S.6
  • 109
    • 0015319592 scopus 로고
    • The biologic clock: the mitochondria?
    • Harman, D. (1972). The biologic clock: the mitochondria? J. Am. Geriatr. Soc. 20, 145-147..
    • (1972) J. Am. Geriatr. Soc. , vol.20 , pp. 145-147
    • Harman, D.1
  • 110
    • 78650880194 scopus 로고    scopus 로고
    • Delayed dominant-negative TNF gene therapy halts progressive loss of nigral dopaminergic neurons in a rat model of Parkinson's disease
    • doi: 10.1038/mt.2010.217
    • Harms, A. S., Barnum, C. J., Ruhn, K. A., Varghese, S., Trevino, I., Blesch, A., et al. (2010). Delayed dominant-negative TNF gene therapy halts progressive loss of nigral dopaminergic neurons in a rat model of Parkinson's disease. Mol. Ther. 19, 46-52. doi: 10.1038/mt.2010.217.
    • (2010) Mol. Ther. , vol.19 , pp. 46-52
    • Harms, A.S.1    Barnum, C.J.2    Ruhn, K.A.3    Varghese, S.4    Trevino, I.5    Blesch, A.6
  • 111
    • 65449135642 scopus 로고    scopus 로고
    • Levodopa: past, present, and future
    • doi: 10.1159/000215875
    • Hauser, R. A. (2009). Levodopa: past, present, and future. Eur. Neurol. 62, 1-8. doi: 10.1159/000215875.
    • (2009) Eur. Neurol. , vol.62 , pp. 1-8
    • Hauser, R.A.1
  • 112
    • 79951720949 scopus 로고    scopus 로고
    • Preladenant in patients with Parkinson's disease and motor fluctuations: a phase 2, double-blind, randomised trial
    • doi: 10.1016/S1474-4422(11)70012-6
    • Hauser, R. A., Cantillon, M., Pourcher, E., Micheli, F., Mok, V., Onofrj, M., et al. (2011). Preladenant in patients with Parkinson's disease and motor fluctuations: a phase 2, double-blind, randomised trial. Lancet Neurol. 10, 221-229. doi: 10.1016/S1474-4422(11)70012-6.
    • (2011) Lancet Neurol. , vol.10 , pp. 221-229
    • Hauser, R.A.1    Cantillon, M.2    Pourcher, E.3    Micheli, F.4    Mok, V.5    Onofrj, M.6
  • 113
    • 61449172156 scopus 로고    scopus 로고
    • Study of istradefylline in patients with Parkinson's disease on levodopa with motor fluctuations
    • doi: 10.1002/mds.22095
    • Hauser, R. A., Shulman, L. M., Trugman, J. M., Roberts, J. W., Mori, A., Ballerini, R., et al. (2008). Study of istradefylline in patients with Parkinson's disease on levodopa with motor fluctuations. Mov. Disord. 23, 2177-2185. doi: 10.1002/mds.22095.
    • (2008) Mov. Disord. , vol.23 , pp. 2177-2185
    • Hauser, R.A.1    Shulman, L.M.2    Trugman, J.M.3    Roberts, J.W.4    Mori, A.5    Ballerini, R.6
  • 114
    • 0032103897 scopus 로고    scopus 로고
    • Characterization of enhanced behavioral responses to L-DOPA following repeated administration in the 6-hydroxydopamine-lesioned rat model of Parkinson's disease
    • doi: 10.1006/exnr.1998.6819
    • Henry, B., Crossman, A. R., and Brotchie, J. M. (1998). Characterization of enhanced behavioral responses to L-DOPA following repeated administration in the 6-hydroxydopamine-lesioned rat model of Parkinson's disease. Exp. Neurol. 151, 334-342. doi: 10.1006/exnr.1998.6819.
    • (1998) Exp. Neurol. , vol.151 , pp. 334-342
    • Henry, B.1    Crossman, A.R.2    Brotchie, J.M.3
  • 115
    • 0032588996 scopus 로고    scopus 로고
    • The alpha2-adrenergic receptor antagonist idazoxan reduces dyskinesia and enhances anti-parkinsonian actions of L-dopa in the MPTP-lesioned primate model of Parkinson's disease
    • Henry, B., Fox, S. H., Peggs, D., Crossman, A. R., and Brotchie, J. M. (1999). The alpha2-adrenergic receptor antagonist idazoxan reduces dyskinesia and enhances anti-parkinsonian actions of L-dopa in the MPTP-lesioned primate model of Parkinson's disease. Mov. Disord. 14, 744-753..
    • (1999) Mov. Disord. , vol.14 , pp. 744-753
    • Henry, B.1    Fox, S.H.2    Peggs, D.3    Crossman, A.R.4    Brotchie, J.M.5
  • 116
    • 34447572589 scopus 로고    scopus 로고
    • Striatal delivery of CERE-120, an AAV2 vector encoding human neurturin, enhances activity of the dopaminergic nigrostriatal system in aged monkeys
    • doi: 10.1002/mds.21503
    • Herzog, C. D., Dass, B., Holden, J. E., Stansell, J. 3rd. Gasmi, M., Tuszynski, M. H., et al. (2007). Striatal delivery of CERE-120, an AAV2 vector encoding human neurturin, enhances activity of the dopaminergic nigrostriatal system in aged monkeys. Mov. Disord. 22, 1124-1132. doi: 10.1002/mds.21503.
    • (2007) Mov. Disord. , vol.22 , pp. 1124-1132
    • Herzog, C.D.1    Dass, B.2    Holden, J.E.3    Stansell, J.4    Gasmi, M.5    Tuszynski, M.H.6
  • 117
    • 0035848459 scopus 로고    scopus 로고
    • Ultrastructural localization of adenosine A2A receptors suggests multiple cellular sites for modulation of GABAergic neurons in rat striatum
    • doi: 10.1002/1096-9861(20010312)431:3%3C331::AID-CNE1074%3E3.0.CO;2-W
    • Hettinger, B. D., Lee, A., Linden, J., and Rosin, D. L. (2001). Ultrastructural localization of adenosine A2A receptors suggests multiple cellular sites for modulation of GABAergic neurons in rat striatum. J. Comp. Neurol. 431, 331-346. doi: 10.1002/1096-9861(20010312)431:3%3C331::AID-CNE1074%3E3.0.CO;2-W.
    • (2001) J. Comp. Neurol. , vol.431 , pp. 331-346
    • Hettinger, B.D.1    Lee, A.2    Linden, J.3    Rosin, D.L.4
  • 118
    • 0032706790 scopus 로고    scopus 로고
    • The adrenergic receptor agonist, clonidine, potentiates the anti-parkinsonian action of the selective kappa-opioid receptor agonist, enadoline, in the monoamine-depleted rat
    • doi: 10.1038/sj.bjp.0702943
    • Hill, M. P., and Brotchie, J. M. (1999). The adrenergic receptor agonist, clonidine, potentiates the anti-parkinsonian action of the selective kappa-opioid receptor agonist, enadoline, in the monoamine-depleted rat. Br. J. Pharmacol. 128, 1577-1585. doi: 10.1038/sj.bjp.0702943.
    • (1999) Br. J. Pharmacol. , vol.128 , pp. 1577-1585
    • Hill, M.P.1    Brotchie, J.M.2
  • 119
    • 84858667311 scopus 로고    scopus 로고
    • Neuroinflammation in Parkinson's disease
    • doi: 10.1016/S1353-8020(11)70065-7
    • Hirsch, E. C., Vyas, S., and Hunot, S. (2012). Neuroinflammation in Parkinson's disease. Parkinsonism. Relat. Disord. 18(Suppl. 1), S210-S212. doi: 10.1016/S1353-8020(11)70065-7.
    • (2012) Parkinsonism. Relat. Disord. , vol.18 , Issue.SUPPL. 1
    • Hirsch, E.C.1    Vyas, S.2    Hunot, S.3
  • 120
    • 67649836782 scopus 로고    scopus 로고
    • Characterization of the potent and highly selective A2A receptor antagonists preladenant and SCH 412348 [7-[2-[4-2,4-difluorophenyl]-1-piperazinyl]ethyl]-2-(2-furanyl)-7H-pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidin-5-amine] in rodent models of movement disorders and depression
    • doi: 10.1124/jpet.108.149617
    • Hodgson, R. A., Bertorelli, R., Varty, G. B., Lachowicz, J. E., Forlani, A., Fredduzzi, S., et al. (2009). Characterization of the potent and highly selective A2A receptor antagonists preladenant and SCH 412348 [7-[2-[4-2,4-difluorophenyl]-1-piperazinyl]ethyl]-2-(2-furanyl)-7H-pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidin-5-amine] in rodent models of movement disorders and depression. J. Pharmacol. Exp. Ther. 330, 294-303. doi: 10.1124/jpet.108.149617.
    • (2009) J. Pharmacol. Exp. Ther. , vol.330 , pp. 294-303
    • Hodgson, R.A.1    Bertorelli, R.2    Varty, G.B.3    Lachowicz, J.E.4    Forlani, A.5    Fredduzzi, S.6
  • 121
    • 0028278560 scopus 로고
    • Cloned glutamate receptors
    • doi: 10.1146/annurev.ne.17.030194.000335
    • Hollmann, M., and Heinemann, S. (1994). Cloned glutamate receptors. Annu. Rev. Neurosci. 17, 31-108. doi: 10.1146/annurev.ne.17.030194.000335.
    • (1994) Annu. Rev. Neurosci. , vol.17 , pp. 31-108
    • Hollmann, M.1    Heinemann, S.2
  • 122
    • 24944458193 scopus 로고    scopus 로고
    • Adenosine A2A, but not A1, receptors mediate the arousal effect of caffeine
    • doi: 10.1038/nn1491
    • Huang, Z. L., Qu, W. M., Eguchi, N., Chen, J. F., Schwarzschild, M. A., Fredholm, B. B., et al. (2005). Adenosine A2A, but not A1, receptors mediate the arousal effect of caffeine. Nat. Neurosci. 8, 858-859. doi: 10.1038/nn1491.
    • (2005) Nat. Neurosci. , vol.8 , pp. 858-859
    • Huang, Z.L.1    Qu, W.M.2    Eguchi, N.3    Chen, J.F.4    Schwarzschild, M.A.5    Fredholm, B.B.6
  • 123
    • 84898612912 scopus 로고    scopus 로고
    • UWA-121, a mixed dopamine serotonin re-uptake inhibitor, enhances l-DOPA anti-parkinsonian action without worsening dyskinesia or psychosis-like behaviours in the MPTP-lesioned common marmoset.
    • doi: 10.1016/j.neuropharm.2014.01.012
    • Huot, P., Johnston, T. H., Lewis, K. D., Koprich, J. B., Reyes, M. G., Fox, S. H., et al. (2014). UWA-121, a mixed dopamine and serotonin re-uptake inhibitor, enhances l-DOPA anti-parkinsonian action without worsening dyskinesia or psychosis-like behaviours in the MPTP-lesioned common marmoset. Neuropharmacology 82C, 76-87. doi: 10.1016/j.neuropharm.2014.01.012.
    • (2014) Neuropharmacology , vol.82 , Issue.C , pp. 76-87
    • Huot, P.1    Johnston, T.H.2    Lewis, K.D.3    Koprich, J.B.4    Reyes, M.G.5    Fox, S.H.6
  • 124
    • 0036020950 scopus 로고    scopus 로고
    • Neuroprotection by adenosine A2A receptor blockade in experimental models of Parkinson's disease
    • doi: 10.1046/j.0022-3042.2001.00694.x
    • Ikeda, K., Kurokawa, M., Aoyama, S., and Kuwana, Y. (2002). Neuroprotection by adenosine A2A receptor blockade in experimental models of Parkinson's disease. J. Neurochem. 80, 262-270. doi: 10.1046/j.0022-3042.2001.00694.x.
    • (2002) J. Neurochem. , vol.80 , pp. 262-270
    • Ikeda, K.1    Kurokawa, M.2    Aoyama, S.3    Kuwana, Y.4
  • 125
    • 79958204001 scopus 로고    scopus 로고
    • The L-type channel antagonist isradipine is neuroprotective in a mouse model of Parkinson's disease
    • doi: 10.1016/j.nbd.2011.04.007
    • Ilijic, E., Guzman, J. N., and Surmeier, D. J. (2011). The L-type channel antagonist isradipine is neuroprotective in a mouse model of Parkinson's disease. Neurobiol. Dis. 43, 364-371. doi: 10.1016/j.nbd.2011.04.007.
    • (2011) Neurobiol. Dis. , vol.43 , pp. 364-371
    • Ilijic, E.1    Guzman, J.N.2    Surmeier, D.J.3
  • 126
    • 0038389063 scopus 로고    scopus 로고
    • The alpha 2-adrenoceptor antagonist idazoxan reverses catalepsy induced by haloperidol in rats independent of striatal dopamine release: role of serotonergic mechanisms
    • doi: 10.1038/sj.npp.13001191300119
    • Invernizzi, R. W., Garavaglia, C., and Samanin, R. (2003). The alpha 2-adrenoceptor antagonist idazoxan reverses catalepsy induced by haloperidol in rats independent of striatal dopamine release: role of serotonergic mechanisms. Neuropsychopharmacology 28, 872-879. doi: 10.1038/sj.npp.13001191300119.
    • (2003) Neuropsychopharmacology , vol.28 , pp. 872-879
    • Invernizzi, R.W.1    Garavaglia, C.2    Samanin, R.3
  • 127
    • 10844220706 scopus 로고    scopus 로고
    • First visualization of adenosine A(2A) receptors in the human brain by positron emission tomography with [11C]TMSX
    • doi: 10.1002/syn.20099
    • Ishiwata, K., Mishina, M., Kimura, Y., Oda, K., Sasaki, T., and Ishii, K. (2005). First visualization of adenosine A(2A) receptors in the human brain by positron emission tomography with [11C]TMSX. Synapse 55, 133-136. doi: 10.1002/syn.20099.
    • (2005) Synapse , vol.55 , pp. 133-136
    • Ishiwata, K.1    Mishina, M.2    Kimura, Y.3    Oda, K.4    Sasaki, T.5    Ishii, K.6
  • 128
    • 84858648303 scopus 로고    scopus 로고
    • Animal models of Parkinson's disease.
    • doi: 10.1016/S1353-8020(11)70057-8
    • Jackson-Lewis, V., Blesa, J., and Przedborski, S. (2012). Animal models of Parkinson's disease. Parkinsonism. Relat. Disord. 18 (Suppl. 1), S183-S185. doi: 10.1016/S1353-8020(11)70057-8.
    • (2012) Parkinsonism. Relat. Disord. , vol.18 , Issue.SUPPL. 1
    • Jackson-Lewis, V.1    Blesa, J.2    Przedborski, S.3
  • 129
    • 68749116710 scopus 로고    scopus 로고
    • A critical evaluation of current staging of alpha-synuclein pathology in Lewy body disorders
    • doi: 10.1016/j.bbadis.2008.07.006
    • Jellinger, K. A. (2009). A critical evaluation of current staging of alpha-synuclein pathology in Lewy body disorders. Biochim. Biophys. Acta 1792, 730-740. doi: 10.1016/j.bbadis.2008.07.006.
    • (2009) Biochim. Biophys. Acta , vol.1792 , pp. 730-740
    • Jellinger, K.A.1
  • 130
    • 67449085790 scopus 로고    scopus 로고
    • Adenosine, adenosine A 2A antagonists, and Parkinson's disease
    • doi: 10.1016/j.parkreldis.2008.12.006
    • Jenner, P., Mori, A., Hauser, R., Morelli, M., Fredholm, B. B., and Chen, J. F. (2009). Adenosine, adenosine A 2A antagonists, and Parkinson's disease. Parkinsonism. Relat. Disord. 15, 406-413. doi: 10.1016/j.parkreldis.2008.12.006.
    • (2009) Parkinsonism. Relat. Disord. , vol.15 , pp. 406-413
    • Jenner, P.1    Mori, A.2    Hauser, R.3    Morelli, M.4    Fredholm, B.B.5    Chen, J.F.6
  • 131
    • 66149086838 scopus 로고    scopus 로고
    • Clinically relevant effects of convection-enhanced delivery of AAV2-GDNF on the dopaminergic nigrostriatal pathway in aged rhesus monkeys
    • doi: 10.1089/hum.2008.137
    • Johnston, L. C., Eberling, J., Pivirotto, P., Hadaczek, P., Federoff, H. J., Forsayeth, J., et al. (2009). Clinically relevant effects of convection-enhanced delivery of AAV2-GDNF on the dopaminergic nigrostriatal pathway in aged rhesus monkeys. Hum. Gene. Ther. 20, 497-510. doi: 10.1089/hum.2008.137.
    • (2009) Hum. Gene. Ther. , vol.20 , pp. 497-510
    • Johnston, L.C.1    Eberling, J.2    Pivirotto, P.3    Hadaczek, P.4    Federoff, H.J.5    Forsayeth, J.6
  • 132
    • 78649377976 scopus 로고    scopus 로고
    • The alpha(2) adrenergic antagonist fipamezole improves quality of levodopa action in Parkinsonian primates
    • doi: 10.1002/mds.23172
    • Johnston, T. H., Fox, S. H., Piggott, M. J., Savola, J. M., and Brotchie, J. M. (2010). The alpha(2) adrenergic antagonist fipamezole improves quality of levodopa action in Parkinsonian primates. Mov. Disord. 25, 2084-2093. doi: 10.1002/mds.23172.
    • (2010) Mov. Disord. , vol.25 , pp. 2084-2093
    • Johnston, T.H.1    Fox, S.H.2    Piggott, M.J.3    Savola, J.M.4    Brotchie, J.M.5
  • 133
    • 77952545692 scopus 로고    scopus 로고
    • Caffeine protects against combined paraquat and maneb-induced dopaminergic neuron degeneration
    • doi: 10.1016/j.expneurol.2010.02.007
    • Kachroo, A., Irizarry, M. C., and Schwarzschild, M. A. (2010). Caffeine protects against combined paraquat and maneb-induced dopaminergic neuron degeneration. Exp. Neurol. 223, 657-661. doi: 10.1016/j.expneurol.2010.02.007.
    • (2010) Exp. Neurol. , vol.223 , pp. 657-661
    • Kachroo, A.1    Irizarry, M.C.2    Schwarzschild, M.A.3
  • 134
    • 27744450594 scopus 로고    scopus 로고
    • Interactions between metabotropic glutamate 5 and adenosine A2A receptors in normal and parkinsonian mice
    • doi: 10.1523/JNEUROSCI.3660-05.2005
    • Kachroo, A., Orlando, L. R., Grandy, D. K., Chen, J. F., Young, A. B., and Schwarzschild, M. A. (2005). Interactions between metabotropic glutamate 5 and adenosine A2A receptors in normal and parkinsonian mice. J. Neurosci. 25, 10414-10419. doi: 10.1523/JNEUROSCI.3660-05.2005.
    • (2005) J. Neurosci. , vol.25 , pp. 10414-10419
    • Kachroo, A.1    Orlando, L.R.2    Grandy, D.K.3    Chen, J.F.4    Young, A.B.5    Schwarzschild, M.A.6
  • 135
    • 33750487494 scopus 로고    scopus 로고
    • Novel neuroprotection by caffeine and adenosine A(2A) receptor antagonists in animal models of Parkinson's disease
    • doi: 10.1016/j.jns.2006.05.003
    • Kalda, A., Yu, L., Oztas, E., and Chen, J. F. (2006). Novel neuroprotection by caffeine and adenosine A(2A) receptor antagonists in animal models of Parkinson's disease. J. Neurol. Sci. 248, 9-15. doi: 10.1016/j.jns.2006.05.003.
    • (2006) J. Neurol. Sci. , vol.248 , pp. 9-15
    • Kalda, A.1    Yu, L.2    Oztas, E.3    Chen, J.F.4
  • 136
    • 0031594271 scopus 로고    scopus 로고
    • Adenosine A2A antagonist: a novel antiparkinsonian agent that does not provoke dyskinesia in parkinsonian monkeys
    • doi: 10.1002/ana.410430415
    • Kanda, T., Jackson, M. J., Smith, L. A., Pearce, R. K., Nakamura, J., Kase, H., et al. (1998). Adenosine A2A antagonist: a novel antiparkinsonian agent that does not provoke dyskinesia in parkinsonian monkeys. Ann. Neurol. 43, 507-513. doi: 10.1002/ana.410430415.
    • (1998) Ann. Neurol. , vol.43 , pp. 507-513
    • Kanda, T.1    Jackson, M.J.2    Smith, L.A.3    Pearce, R.K.4    Nakamura, J.5    Kase, H.6
  • 137
    • 84869409550 scopus 로고    scopus 로고
    • CaV1.3-selective L-type calcium channel antagonists as potential new therapeutics for Parkinson's disease
    • doi: 10.1038/ncomms2149
    • Kang, S., Cooper, G., Dunne, S. F., Dusel, B., Luan, C. H., and Surmeier, D. J. (2012). CaV1.3-selective L-type calcium channel antagonists as potential new therapeutics for Parkinson's disease. Nat. Commun. 3, 1146. doi: 10.1038/ncomms2149.
    • (2012) Nat. Commun. , vol.3 , pp. 1146
    • Kang, S.1    Cooper, G.2    Dunne, S.F.3    Dusel, B.4    Luan, C.H.5    Surmeier, D.J.6
  • 138
    • 0036300978 scopus 로고    scopus 로고
    • [Tandospirone citrate, a selective 5-HT1A agonist, alleviates L-DOPA-induced dyskinesia in patients with Parkinson's disease]
    • Kannari, K., Kurahashi, K., Tomiyama, M., Maeda, T., Arai, A., Baba, M., et al. (2002). [Tandospirone citrate, a selective 5-HT1A agonist, alleviates L-DOPA-induced dyskinesia in patients with Parkinson's disease]. Shinkei. 54, 133-137.
    • (2002) Shinkei. , vol.54 , pp. 133-137
    • Kannari, K.1    Kurahashi, K.2    Tomiyama, M.3    Maeda, T.4    Arai, A.5    Baba, M.6
  • 139
    • 10744230559 scopus 로고    scopus 로고
    • A past psychiatric history may be a risk factor for topiramate-related psychiatric and cognitive adverse events
    • doi: 10.1016/j.yebeh.2003.07.007
    • Kanner, A. M., Wuu, J., Faught, E., Tatum, W. O. T., Fix, A., and French, J. A. (2003). A past psychiatric history may be a risk factor for topiramate-related psychiatric and cognitive adverse events. Epilepsy. Behav. 4, 548-552. doi: 10.1016/j.yebeh.2003.07.007.
    • (2003) Epilepsy. Behav. , vol.4 , pp. 548-552
    • Kanner, A.M.1    Wuu, J.2    Faught, E.3    Tatum, W.O.T.4    Fix, A.5    French, J.A.6
  • 140
    • 34250683023 scopus 로고    scopus 로고
    • Safety and tolerability of gene therapy with an adeno-associated virus (AAV) borne GAD gene for Parkinson's disease: an open label, phase I trial
    • doi: 10.1016/S0140-6736(07)60982-9
    • Kaplitt, M. G., Feigin, A., Tang, C., Fitzsimons, H. L., Mattis, P., Lawlor, P. A., et al. (2007). Safety and tolerability of gene therapy with an adeno-associated virus (AAV) borne GAD gene for Parkinson's disease: an open label, phase I trial. Lancet 369, 2097-2105. doi: 10.1016/S0140-6736(07)60982-9.
    • (2007) Lancet , vol.369 , pp. 2097-2105
    • Kaplitt, M.G.1    Feigin, A.2    Tang, C.3    Fitzsimons, H.L.4    Mattis, P.5    Lawlor, P.A.6
  • 141
    • 0242684415 scopus 로고    scopus 로고
    • Genetic or pharmacological iron chelation prevents MPTP-induced neurotoxicity in vivo: a novel therapy for Parkinson's disease
    • doi: 10.1016/S0896-6273(03)00126-0
    • Kaur, D., Yantiri, F., Rajagopalan, S., Kumar, J., Mo, J. Q., Boonplueang, R., et al. (2003). Genetic or pharmacological iron chelation prevents MPTP-induced neurotoxicity in vivo: a novel therapy for Parkinson's disease. Neuron 37, 899-909. doi: 10.1016/S0896-6273(03)00126-0.
    • (2003) Neuron , vol.37 , pp. 899-909
    • Kaur, D.1    Yantiri, F.2    Rajagopalan, S.3    Kumar, J.4    Mo, J.Q.5    Boonplueang, R.6
  • 142
    • 18544391840 scopus 로고    scopus 로고
    • MK 801 reverses haloperidol-induced catalepsy from both striatal and extrastriatal sites in the rat brain
    • doi: 10.1016/S0014-2999(97)01078-9
    • Kaur, S., Ozer, H., and Starr, M. (1997). MK 801 reverses haloperidol-induced catalepsy from both striatal and extrastriatal sites in the rat brain. Eur. J. Pharmacol. 332, 153-160. doi: 10.1016/S0014-2999(97)01078-9.
    • (1997) Eur. J. Pharmacol. , vol.332 , pp. 153-160
    • Kaur, S.1    Ozer, H.2    Starr, M.3
  • 143
    • 0029063028 scopus 로고
    • Antiparkinsonian action of dextromethorphan in the reserpine-treated mouse
    • doi: 10.1016/0014-2999(95)00200-5
    • Kaur, S., and Starr, M. S. (1995). Antiparkinsonian action of dextromethorphan in the reserpine-treated mouse. Eur. J. Pharmacol. 280, 159-166. doi: 10.1016/0014-2999(95)00200-5.
    • (1995) Eur. J. Pharmacol. , vol.280 , pp. 159-166
    • Kaur, S.1    Starr, M.S.2
  • 144
    • 0032543993 scopus 로고    scopus 로고
    • The role of nigrostriatal dopamine in metabotropic glutamate agonist-induced rotation
    • Kearney, J. A., Becker, J. B., Frey, K. A., and Albin, R. L. (1998). The role of nigrostriatal dopamine in metabotropic glutamate agonist-induced rotation. Neuroscience 87, 881-891..
    • (1998) Neuroscience , vol.87 , pp. 881-891
    • Kearney, J.A.1    Becker, J.B.2    Frey, K.A.3    Albin, R.L.4
  • 145
    • 0001571891 scopus 로고    scopus 로고
    • Metabotropic glutamate agonist-induced rotation: a pharmacological, FOS immunohistochemical, and [14C]-2-deoxyglucose autoradiographic study
    • Kearney, J. A., Frey, K. A., and Albin, R. L. (1997). Metabotropic glutamate agonist-induced rotation: a pharmacological, FOS immunohistochemical, and [14C]-2-deoxyglucose autoradiographic study. J. Neurosci. 17, 4415-4425..
    • (1997) J. Neurosci. , vol.17 , pp. 4415-4425
    • Kearney, J.A.1    Frey, K.A.2    Albin, R.L.3
  • 146
    • 4544357027 scopus 로고    scopus 로고
    • NMDA receptor antagonists ameliorate the stepping deficits produced by unilateral medial forebrain bundle injections of 6-OHDA in rats
    • doi: 10.1007/s00213-004-1799-5
    • Kelsey, J. E., Mague, S. D., Pijanowski, R. S., Harris, R. C., Kleckner, N. W., and Matthews, R. T. (2004). NMDA receptor antagonists ameliorate the stepping deficits produced by unilateral medial forebrain bundle injections of 6-OHDA in rats. Psychopharmacology (Berl.) 175, 179-188. doi: 10.1007/s00213-004-1799-5.
    • (2004) Psychopharmacology (Berl.) , vol.175 , pp. 179-188
    • Kelsey, J.E.1    Mague, S.D.2    Pijanowski, R.S.3    Harris, R.C.4    Kleckner, N.W.5    Matthews, R.T.6
  • 147
    • 59649114125 scopus 로고    scopus 로고
    • Synaptic AMPA receptor plasticity and behavior
    • doi: 10.1016/j.neuron.2009.01.015
    • Kessels, H. W., and Malinow, R. (2009). Synaptic AMPA receptor plasticity and behavior. Neuron 61, 340-350. doi: 10.1016/j.neuron.2009.01.015.
    • (2009) Neuron , vol.61 , pp. 340-350
    • Kessels, H.W.1    Malinow, R.2
  • 148
    • 0033773237 scopus 로고    scopus 로고
    • Preservation of a functional nigrostriatal dopamine pathway by GDNF in the intrastriatal 6-OHDA lesion model depends on the site of administration of the trophic factor.
    • doi: 10.1046/j.1460-9568.2000.00274.x
    • Kirik, D., Rosenblad, C., and Bjorklund, A. (2000a). Preservation of a functional nigrostriatal dopamine pathway by GDNF in the intrastriatal 6-OHDA lesion model depends on the site of administration of the trophic factor. Eur. J. Neurosci. 12, 3871-3882. doi: 10.1046/j.1460-9568.2000.00274.x.
    • (2000) Eur. J. Neurosci. , vol.12 , pp. 3871-3882
    • Kirik, D.1    Rosenblad, C.2    Bjorklund, A.3
  • 149
    • 0034659835 scopus 로고    scopus 로고
    • Long-term rAAV-mediated gene transfer of GDNF in the rat Parkinson's model: intrastriatal but not intranigral transduction promotes functional regeneration in the lesioned nigrostriatal system.
    • Kirik, D., Rosenblad, C., Bjorklund, A., and Mandel, R. J. (2000b). Long-term rAAV-mediated gene transfer of GDNF in the rat Parkinson's model: intrastriatal but not intranigral transduction promotes functional regeneration in the lesioned nigrostriatal system. J. Neurosci. 20, 4686-4700..
    • (2000) J. Neurosci. , vol.20 , pp. 4686-4700
    • Kirik, D.1    Rosenblad, C.2    Bjorklund, A.3    Mandel, R.J.4
  • 150
    • 0037208682 scopus 로고    scopus 로고
    • Biochemistry of Parkinson's disease: is a brain serotonergic deficiency a characteristic of idiopathic Parkinson's disease?
    • Kish, S. J. (2003). Biochemistry of Parkinson's disease: is a brain serotonergic deficiency a characteristic of idiopathic Parkinson's disease? Adv. Neurol. 91, 39-49..
    • (2003) Adv. Neurol. , vol.91 , pp. 39-49
    • Kish, S.J.1
  • 151
    • 84872154697 scopus 로고    scopus 로고
    • Genetics of Parkinson's disease.
    • doi: 10.1101/cshperspect.a008888
    • Klein, C., and Westenberger, A. (2012). Genetics of Parkinson's disease. Cold. Spring. Harb. Perspect. Med. 2:a008888. doi: 10.1101/cshperspect.a008888.
    • (2012) Cold. Spring. Harb. Perspect. Med. , vol.2
    • Klein, C.1    Westenberger, A.2
  • 152
    • 0026074931 scopus 로고
    • The AMPA receptor antagonist NBQX has antiparkinsonian effects in monoamine-depleted rats and MPTP-treated monkeys
    • doi: 10.1002/ana.410300513
    • Klockgether, T., Turski, L., Honore, T., Zhang, Z. M., Gash, D. M., Kurlan, R., et al. (1991). The AMPA receptor antagonist NBQX has antiparkinsonian effects in monoamine-depleted rats and MPTP-treated monkeys. Ann. Neurol. 30, 717-723. doi: 10.1002/ana.410300513.
    • (1991) Ann. Neurol. , vol.30 , pp. 717-723
    • Klockgether, T.1    Turski, L.2    Honore, T.3    Zhang, Z.M.4    Gash, D.M.5    Kurlan, R.6
  • 153
    • 0034520395 scopus 로고    scopus 로고
    • Inflammatory regulators in Parkinson's disease: iNOS, lipocortin-1, and cyclooxygenases-1 and -2
    • doi: 10.1006/mcne.2000.0914
    • Knott, C., Stern, G., and Wilkin, G. P. (2000). Inflammatory regulators in Parkinson's disease: iNOS, lipocortin-1, and cyclooxygenases-1 and -2. Mol. Cell Neurosci. 16, 724-739. doi: 10.1006/mcne.2000.0914.
    • (2000) Mol. Cell Neurosci. , vol.16 , pp. 724-739
    • Knott, C.1    Stern, G.2    Wilkin, G.P.3
  • 154
    • 84897956912 scopus 로고    scopus 로고
    • Randomized clinical trial of topiramate for levodopa-induced dyskinesia in Parkinson's disease
    • doi: 10.1016/j.parkreldis.2014.01.016
    • Kobylecki, C., Burn, D. J., Kass-Iliyya, L., Kellett, M. W., Crossman, A. R., and Silverdale, M. A. (2014). Randomized clinical trial of topiramate for levodopa-induced dyskinesia in Parkinson's disease. Parkinsonism. Relat. Disord. 240, 452-455. doi: 10.1016/j.parkreldis.2014.01.016.
    • (2014) Parkinsonism. Relat. Disord. , vol.240 , pp. 452-455
    • Kobylecki, C.1    Burn, D.J.2    Kass-Iliyya, L.3    Kellett, M.W.4    Crossman, A.R.5    Silverdale, M.A.6
  • 155
    • 81955164826 scopus 로고    scopus 로고
    • Synergistic antidyskinetic effects of topiramate and amantadine in animal models of Parkinson's disease
    • doi: 10.1002/mds.23867
    • Kobylecki, C., Hill, M. P., Crossman, A. R., and Ravenscroft, P. (2011). Synergistic antidyskinetic effects of topiramate and amantadine in animal models of Parkinson's disease. Mov. Disord. 26, 2354-2363. doi: 10.1002/mds.23867.
    • (2011) Mov. Disord. , vol.26 , pp. 2354-2363
    • Kobylecki, C.1    Hill, M.P.2    Crossman, A.R.3    Ravenscroft, P.4
  • 156
    • 0032913134 scopus 로고    scopus 로고
    • L-701,324, a selective antagonist at the glycine site of the NMDA receptor, counteracts haloperidol-induced muscle rigidity in rats
    • doi: 10.1007/s002130050942
    • Konieczny, J., Ossowska, K., Schulze, G., Coper, H., and Wolfarth, S. (1999). L-701,324, a selective antagonist at the glycine site of the NMDA receptor, counteracts haloperidol-induced muscle rigidity in rats. Psychopharmacology (Berl.) 143, 235-243. doi: 10.1007/s002130050942.
    • (1999) Psychopharmacology (Berl.) , vol.143 , pp. 235-243
    • Konieczny, J.1    Ossowska, K.2    Schulze, G.3    Coper, H.4    Wolfarth, S.5
  • 157
    • 0034090891 scopus 로고    scopus 로고
    • AMPA receptor blockade improves levodopa-induced dyskinesia in MPTP monkeys
    • doi: 10.1212/WNL.54.8.1589
    • Konitsiotis, S., Blanchet, P. J., Verhagen, L., Lamers, E., and Chase, T. N. (2000). AMPA receptor blockade improves levodopa-induced dyskinesia in MPTP monkeys. Neurology. 54, 1589-1595. doi: 10.1212/WNL.54.8.1589.
    • (2000) Neurology. , vol.54 , pp. 1589-1595
    • Konitsiotis, S.1    Blanchet, P.J.2    Verhagen, L.3    Lamers, E.4    Chase, T.N.5
  • 158
    • 33845993229 scopus 로고    scopus 로고
    • Delivery of neurturin by AAV2 (CERE-120)-mediated gene transfer provides structural and functional neuroprotection and neurorestoration in MPTP-treated monkeys
    • doi: 10.1002/ana.21032
    • Kordower, J. H., Herzog, C. D., Dass, B., Bakay, R. A., Stansell, J. 3rd. Gasmi, M., et al. (2006). Delivery of neurturin by AAV2 (CERE-120)-mediated gene transfer provides structural and functional neuroprotection and neurorestoration in MPTP-treated monkeys. Ann. Neurol. 60, 706-715. doi: 10.1002/ana.21032.
    • (2006) Ann. Neurol. , vol.60 , pp. 706-715
    • Kordower, J.H.1    Herzog, C.D.2    Dass, B.3    Bakay, R.A.4    Stansell, J.5    Gasmi, M.6
  • 159
    • 0032850453 scopus 로고    scopus 로고
    • Clinicopathological findings following intraventricular glial-derived neurotrophic factor treatment in a patient with Parkinson's disease
    • Kordower, J. H., Palfi, S., Chen, E. Y., Ma, S. Y., Sendera, T., Cochran, E. J., et al. (1999). Clinicopathological findings following intraventricular glial-derived neurotrophic factor treatment in a patient with Parkinson's disease. Ann. Neurol. 46, 419-424..
    • (1999) Ann. Neurol. , vol.46 , pp. 419-424
    • Kordower, J.H.1    Palfi, S.2    Chen, E.Y.3    Ma, S.Y.4    Sendera, T.5    Cochran, E.J.6
  • 160
    • 0029827559 scopus 로고    scopus 로고
    • Neurturin, a relative of glial-cell-line-derived neurotrophic factor
    • doi: 10.1038/384467a0
    • Kotzbauer, P. T., Lampe, P. A., Heuckeroth, R. O., Golden, J. P., Creedon, D. J., Johnson, E. M. Jr., et al. (1996). Neurturin, a relative of glial-cell-line-derived neurotrophic factor. Nature 384, 467-470. doi: 10.1038/384467a0.
    • (1996) Nature , vol.384 , pp. 467-470
    • Kotzbauer, P.T.1    Lampe, P.A.2    Heuckeroth, R.O.3    Golden, J.P.4    Creedon, D.J.5    Johnson Jr., E.M.6
  • 161
    • 38949126940 scopus 로고    scopus 로고
    • Excitotoxicity and new antiglutamatergic strategies in Parkinson's disease and Alzheimer's disease
    • doi: 10.1016/S1353-8020(08)70025-7
    • Koutsilieri, E., and Riederer, P. (2007). Excitotoxicity and new antiglutamatergic strategies in Parkinson's disease and Alzheimer's disease. Parkinsonism. Relat. Disord. 13(Suppl. 3), S329-S331. doi: 10.1016/S1353-8020(08)70025-7.
    • (2007) Parkinsonism. Relat. Disord. , vol.13 , Issue.SUPPL. 3
    • Koutsilieri, E.1    Riederer, P.2
  • 162
    • 77955172369 scopus 로고    scopus 로고
    • Regulation of parkinsonian motor behaviours by optogenetic control of basal ganglia circuitry
    • doi: 10.1038/nature09159
    • Kravitz, A. V., Freeze, B. S., Parker, P. R., Kay, K., Thwin, M. T., Deisseroth, K., et al. (2010). Regulation of parkinsonian motor behaviours by optogenetic control of basal ganglia circuitry. Nature 466, 622-626. doi: 10.1038/nature09159.
    • (2010) Nature , vol.466 , pp. 622-626
    • Kravitz, A.V.1    Freeze, B.S.2    Parker, P.R.3    Kay, K.4    Thwin, M.T.5    Deisseroth, K.6
  • 163
    • 0031721139 scopus 로고    scopus 로고
    • Double-blind evaluation of subthalamic nucleus deep brain stimulation in advanced Parkinson's disease
    • doi: 10.1212/WNL.51.3.850
    • Kumar, R., Lozano, A. M., Kim, Y. J., Hutchison, W. D., Sime, E., Halket, E., et al. (1998). Double-blind evaluation of subthalamic nucleus deep brain stimulation in advanced Parkinson's disease. Neurology 51, 850-855. doi: 10.1212/WNL.51.3.850.
    • (1998) Neurology , vol.51 , pp. 850-855
    • Kumar, R.1    Lozano, A.M.2    Kim, Y.J.3    Hutchison, W.D.4    Sime, E.5    Halket, E.6
  • 165
    • 34249992252 scopus 로고    scopus 로고
    • Localization and expression of group I metabotropic glutamate receptors in the mouse striatum, globus pallidus, and subthalamic nucleus: regulatory effects of MPTP treatment and constitutive Homer deletion
    • doi: 10.1523/JNEUROSCI.3819-06.2007
    • Kuwajima, M., Dehoff, M. H., Furuichi, T., Worley, P. F., Hall, R. A., and Smith, Y. (2007). Localization and expression of group I metabotropic glutamate receptors in the mouse striatum, globus pallidus, and subthalamic nucleus: regulatory effects of MPTP treatment and constitutive Homer deletion. J. Neurosci. 27, 6249-6260. doi: 10.1523/JNEUROSCI.3819-06.2007.
    • (2007) J. Neurosci. , vol.27 , pp. 6249-6260
    • Kuwajima, M.1    Dehoff, M.H.2    Furuichi, T.3    Worley, P.F.4    Hall, R.A.5    Smith, Y.6
  • 166
    • 77954649301 scopus 로고    scopus 로고
    • Kyowa Hakko Receives Not Approvable Letter from FDA for Istradefylline (KW-6002) [Media Release].
    • Kyowa Hakko Kirin Co Ltd., Available online at:, [Accessed 10 December 2013]
    • Kyowa Hakko Kirin Co Ltd. (2008). Kyowa Hakko Receives Not Approvable Letter from FDA for Istradefylline (KW-6002) [Media Release]. Available online at: http://www.kyowa-kirin.com/news_releases/kyowa/2008/er080228_01.html [Accessed 10 December 2013].
    • (2008)
  • 167
    • 84907178116 scopus 로고    scopus 로고
    • Approval for Manufacturing Marketing of NOURIAST Tablets 20 mg, a Novel Antiparkinsonian Agent [Media Release].
    • Kyowa Hakko Kirin Co Ltd., Available online:, [Accessed 10 December 2013]
    • Kyowa Hakko Kirin Co Ltd. (2013). Approval for Manufacturing and Marketing of NOURIAST Tablets 20 mg, a Novel Antiparkinsonian Agent [Media Release]. Available online: http://www.kyowa-kirin.com/news_releases/2013/e20130325_04.html [Accessed 10 December 2013].
    • (2013)
  • 168
    • 0030864433 scopus 로고    scopus 로고
    • Desferrioxamine and vitamin E protect against iron and MPTP-induced neurodegeneration in mice
    • doi: 10.1007/BF01277665
    • Lan, J., and Jiang, D. H. (1997). Desferrioxamine and vitamin E protect against iron and MPTP-induced neurodegeneration in mice. J. Neural. Transm. 104, 469-481. doi: 10.1007/BF01277665.
    • (1997) J. Neural. Transm. , vol.104 , pp. 469-481
    • Lan, J.1    Jiang, D.H.2
  • 169
    • 0027765719 scopus 로고
    • The competitive NMDA antagonist CPP protects substantia nigra neurons from MPTP-induced degeneration in primates
    • doi: 10.1007/BF00167234
    • Lange, K. W., Loschmann, P. A., Sofic, E., Burg, M., Horowski, R., Kalveram, K. T., et al. (1993). The competitive NMDA antagonist CPP protects substantia nigra neurons from MPTP-induced degeneration in primates. Naunyn. Schmiedebergs. Arch. Pharmacol. 348, 586-592. doi: 10.1007/BF00167234.
    • (1993) Naunyn. Schmiedebergs. Arch. Pharmacol. , vol.348 , pp. 586-592
    • Lange, K.W.1    Loschmann, P.A.2    Sofic, E.3    Burg, M.4    Horowski, R.5    Kalveram, K.T.6
  • 170
    • 0034765853 scopus 로고    scopus 로고
    • Adenosine in the central nervous system: release mechanisms and extracellular concentrations
    • doi: 10.1046/j.1471-4159.2001.00607.x
    • Latini, S., and Pedata, F. (2001). Adenosine in the central nervous system: release mechanisms and extracellular concentrations. J. Neurochem 79, 463-484. doi: 10.1046/j.1471-4159.2001.00607.x.
    • (2001) J. Neurochem , vol.79 , pp. 463-484
    • Latini, S.1    Pedata, F.2
  • 171
    • 34249099711 scopus 로고    scopus 로고
    • NMDA receptor trafficking in synaptic plasticity and neuropsychiatric disorders
    • doi: 10.1038/nrn2153
    • Lau, C. G., and Zukin, R. S. (2007). NMDA receptor trafficking in synaptic plasticity and neuropsychiatric disorders. Nat. Rev. Neurosci. 8, 413-426. doi: 10.1038/nrn2153.
    • (2007) Nat. Rev. Neurosci. , vol.8 , pp. 413-426
    • Lau, C.G.1    Zukin, R.S.2
  • 172
    • 0025050481 scopus 로고
    • Immunohistochemical study of the serotoninergic innervation of the basal ganglia in the squirrel monkey
    • doi: 10.1002/cne.902990102
    • Lavoie, B., and Parent, A. (1990). Immunohistochemical study of the serotoninergic innervation of the basal ganglia in the squirrel monkey. J. Comp. Neurol. 299, 1-16. doi: 10.1002/cne.902990102.
    • (1990) J. Comp. Neurol. , vol.299 , pp. 1-16
    • Lavoie, B.1    Parent, A.2
  • 173
    • 23744497262 scopus 로고    scopus 로고
    • Enhanced expression of glutamate decarboxylase 65 improves symptoms of rat parkinsonian models
    • doi: 10.1038/sj.gt.3302520
    • Lee, B., Lee, H., Nam, Y. R., Oh, J. H., Cho, Y. H., and Chang, J. W. (2005). Enhanced expression of glutamate decarboxylase 65 improves symptoms of rat parkinsonian models. Gene. Ther. 12, 1215-1222. doi: 10.1038/sj.gt.3302520.
    • (2005) Gene. Ther. , vol.12 , pp. 1215-1222
    • Lee, B.1    Lee, H.2    Nam, Y.R.3    Oh, J.H.4    Cho, Y.H.5    Chang, J.W.6
  • 174
    • 84856952320 scopus 로고    scopus 로고
    • Perampanel, an AMPA antagonist, found to have no benefit in reducing "off" time in Parkinson's disease
    • doi: 10.1002/mds.23983
    • Lees, A., Fahn, S., Eggert, K. M., Jankovic, J., Lang, A., Micheli, F., et al. (2012). Perampanel, an AMPA antagonist, found to have no benefit in reducing "off" time in Parkinson's disease. Mov. Disord. 27, 284-288. doi: 10.1002/mds.23983.
    • (2012) Mov. Disord. , vol.27 , pp. 284-288
    • Lees, A.1    Fahn, S.2    Eggert, K.M.3    Jankovic, J.4    Lang, A.5    Micheli, F.6
  • 175
    • 0033067301 scopus 로고    scopus 로고
    • Long-term restoration of striatal L-aromatic amino acid decarboxylase activity using recombinant adeno-associated viral vector gene transfer in a rodent model of Parkinson's disease
    • doi: 10.1016/S0306-4522(98)00741-6
    • Leff, S. E., Spratt, S. K., Snyder, R. O., and Mandel, R. J. (1999). Long-term restoration of striatal L-aromatic amino acid decarboxylase activity using recombinant adeno-associated viral vector gene transfer in a rodent model of Parkinson's disease. Neuroscience 92, 185-196. doi: 10.1016/S0306-4522(98)00741-6.
    • (1999) Neuroscience , vol.92 , pp. 185-196
    • Leff, S.E.1    Spratt, S.K.2    Snyder, R.O.3    Mandel, R.J.4
  • 176
    • 41849131038 scopus 로고    scopus 로고
    • Adenosine A2A receptor antagonist istradefylline (KW-6002) reduces "off" time in Parkinson's disease: a double-blind, randomized, multicenter clinical trial (6002-US-005)
    • doi: 10.1002/ana.21315
    • Lewitt, P. A., Guttman, M., Tetrud, J. W., Tuite, P. J., Mori, A., Chaikin, P., et al. (2008). Adenosine A2A receptor antagonist istradefylline (KW-6002) reduces "off" time in Parkinson's disease: a double-blind, randomized, multicenter clinical trial (6002-US-005). Ann. Neurol. 63, 295-302. doi: 10.1002/ana.21315.
    • (2008) Ann. Neurol. , vol.63 , pp. 295-302
    • Lewitt, P.A.1    Guttman, M.2    Tetrud, J.W.3    Tuite, P.J.4    Mori, A.5    Chaikin, P.6
  • 177
    • 84866249555 scopus 로고    scopus 로고
    • Randomized clinical trial of fipamezole for dyskinesia in Parkinson disease (FJORD study)
    • doi: 10.1212/WNL.0b013e31825f0451
    • Lewitt, P. A., Hauser, R. A., Lu, M., Nicholas, A. P., Weiner, W., Coppard, N., et al. (2012). Randomized clinical trial of fipamezole for dyskinesia in Parkinson disease (FJORD study). Neurology 79, 163-169. doi: 10.1212/WNL.0b013e31825f0451.
    • (2012) Neurology , vol.79 , pp. 163-169
    • Lewitt, P.A.1    Hauser, R.A.2    Lu, M.3    Nicholas, A.P.4    Weiner, W.5    Coppard, N.6
  • 178
    • 84886413067 scopus 로고    scopus 로고
    • 3-hydroxykynurenine and other Parkinson's disease biomarkers discovered by metabolomic analysis
    • doi: 10.1002/mds.25555
    • Lewitt, P. A., Li, J., Lu, M., Beach, T. G., Adler, C. H., and Guo, L. (2013). 3-hydroxykynurenine and other Parkinson's disease biomarkers discovered by metabolomic analysis. Mov. Disord. 28, 1653-1660. doi: 10.1002/mds.25555.
    • (2013) Mov. Disord. , vol.28 , pp. 1653-1660
    • Lewitt, P.A.1    Li, J.2    Lu, M.3    Beach, T.G.4    Adler, C.H.5    Guo, L.6
  • 179
    • 79952740079 scopus 로고    scopus 로고
    • AAV2-GAD gene therapy for advanced Parkinson's disease: a double-blind, sham-surgery controlled, randomised trial
    • doi: 10.1016/S1474-4422(11)70039-4
    • Lewitt, P. A., Rezai, A. R., Leehey, M. A., Ojemann, S. G., Flaherty, A. W., Eskandar, E. N., et al. (2011). AAV2-GAD gene therapy for advanced Parkinson's disease: a double-blind, sham-surgery controlled, randomised trial. Lancet Neurol. 10, 309-319. doi: 10.1016/S1474-4422(11)70039-4.
    • (2011) Lancet Neurol. , vol.10 , pp. 309-319
    • Lewitt, P.A.1    Rezai, A.R.2    Leehey, M.A.3    Ojemann, S.G.4    Flaherty, A.W.5    Eskandar, E.N.6
  • 180
    • 80051660738 scopus 로고    scopus 로고
    • Iron chelation and neuroprotection in neurodegenerative diseases
    • doi: 10.1007/s00702-010-0518-0
    • Li, X., Jankovic, J., and Le, W. (2011). Iron chelation and neuroprotection in neurodegenerative diseases. J. Neural. Transm. 118, 473-477. doi: 10.1007/s00702-010-0518-0.
    • (2011) J. Neural. Transm. , vol.118 , pp. 473-477
    • Li, X.1    Jankovic, J.2    Le, W.3
  • 181
    • 59049087723 scopus 로고    scopus 로고
    • GLP-1 receptor stimulation preserves primary cortical and dopaminergic neurons in cellular and rodent models of stroke and Parkinsonism
    • doi: 10.1073/pnas.0806720106
    • Li, Y., Perry, T., Kindy, M. S., Harvey, B. K., Tweedie, D., Holloway, H. W., et al. (2009). GLP-1 receptor stimulation preserves primary cortical and dopaminergic neurons in cellular and rodent models of stroke and Parkinsonism. Proc. Natl. Acad. Sci. U.S.A. 106, 1285-1290. doi: 10.1073/pnas.0806720106.
    • (2009) Proc. Natl. Acad. Sci. U.S.A. , vol.106 , pp. 1285-1290
    • Li, Y.1    Perry, T.2    Kindy, M.S.3    Harvey, B.K.4    Tweedie, D.5    Holloway, H.W.6
  • 182
    • 0027285510 scopus 로고
    • GDNF: a glial cell line-derived neurotrophic factor for midbrain dopaminergic neurons
    • doi: 10.1126/science.8493557
    • Lin, L. F., Doherty, D. H., Lile, J. D., Bektesh, S., and Collins, F. (1993). GDNF: a glial cell line-derived neurotrophic factor for midbrain dopaminergic neurons. Science 260, 1130-1132. doi: 10.1126/science.8493557.
    • (1993) Science , vol.260 , pp. 1130-1132
    • Lin, L.F.1    Doherty, D.H.2    Lile, J.D.3    Bektesh, S.4    Collins, F.5
  • 183
    • 84894738679 scopus 로고    scopus 로고
    • In vivo detection of monoaminergic degeneration in early Parkinson disease by 18F-9-Fluoropropyl-(+)-Dihydrotetrabenzazine PET
    • doi: 10.2967/jnumed.113.121897
    • Lin, S. C., Lin, K. J., Hsiao, I. T., Hsieh, C. J., Lin, W. Y., Lu, C. S., et al. (2013). In vivo detection of monoaminergic degeneration in early Parkinson disease by 18F-9-Fluoropropyl-(+)-Dihydrotetrabenzazine PET. J. Nucl. Med. 55, 73-79. doi: 10.2967/jnumed.113.121897.
    • (2013) J. Nucl. Med. , vol.55 , pp. 73-79
    • Lin, S.C.1    Lin, K.J.2    Hsiao, I.T.3    Hsieh, C.J.4    Lin, W.Y.5    Lu, C.S.6
  • 184
    • 77349086589 scopus 로고    scopus 로고
    • L-DOPA-induced dopamine efflux in the striatum and the substantia nigra in a rat model of Parkinson's disease: temporal and quantitative relationship to the expression of dyskinesia
    • doi: 10.1111/j.1471-4159.2009.06556.x
    • Lindgren, H. S., Andersson, D. R., Lagerkvist, S., Nissbrandt, H., and Cenci, M. A. (2010). L-DOPA-induced dopamine efflux in the striatum and the substantia nigra in a rat model of Parkinson's disease: temporal and quantitative relationship to the expression of dyskinesia. J. Neurochem. 112, 1465-1476. doi: 10.1111/j.1471-4159.2009.06556.x.
    • (2010) J. Neurochem. , vol.112 , pp. 1465-1476
    • Lindgren, H.S.1    Andersson, D.R.2    Lagerkvist, S.3    Nissbrandt, H.4    Cenci, M.A.5
  • 185
    • 13944261368 scopus 로고    scopus 로고
    • L-type calcium channels: the low down
    • doi: 10.1152/jn.00486.2004
    • Lipscombe, D., Helton, T. D., and Xu, W. (2004). L-type calcium channels: the low down. J. Neurophysiol. 92, 2633-2641. doi: 10.1152/jn.00486.2004.
    • (2004) J. Neurophysiol. , vol.92 , pp. 2633-2641
    • Lipscombe, D.1    Helton, T.D.2    Xu, W.3
  • 186
    • 0027008315 scopus 로고
    • Synergism of NBQX with dopamine agonists in the 6-OHDA rat model of Parkinson's disease
    • Loschmann, P. A., Kunow, M., and Wachtel, H. (1992). Synergism of NBQX with dopamine agonists in the 6-OHDA rat model of Parkinson's disease. J. Neural. Transm. Suppl. 38, 55-64..
    • (1992) J. Neural. Transm. Suppl. , vol.38 , pp. 55-64
    • Loschmann, P.A.1    Kunow, M.2    Wachtel, H.3
  • 187
    • 0025992357 scopus 로고
    • Synergism of the AMPA-antagonist NBQX and the NMDA-antagonist CPP with L-dopa in models of Parkinson's disease
    • doi: 10.1007/BF02259538
    • Loschmann, P. A., Lange, K. W., Kunow, M., Rettig, K. J., Jahnig, P., Honore, T., et al. (1991). Synergism of the AMPA-antagonist NBQX and the NMDA-antagonist CPP with L-dopa in models of Parkinson's disease. J. Neural. Transm. Park. Dis. Dement. Sect. 3, 203-213. doi: 10.1007/BF02259538.
    • (1991) J. Neural. Transm. Park. Dis. Dement. Sect. , vol.3 , pp. 203-213
    • Loschmann, P.A.1    Lange, K.W.2    Kunow, M.3    Rettig, K.J.4    Jahnig, P.5    Honore, T.6
  • 188
    • 84869109864 scopus 로고    scopus 로고
    • Pathological alpha-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice
    • doi: 10.1126/science.1227157
    • Luk, K. C., Kehm, V., Carroll, J., Zhang, B., O'Brien, P., and Trojanowski, J. Q. (2012). Pathological alpha-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science 338, 949-953. doi: 10.1126/science.1227157.
    • (2012) Science , vol.338 , pp. 949-953
    • Luk, K.C.1    Kehm, V.2    Carroll, J.3    Zhang, B.4    O'Brien, P.5    Trojanowski, J.Q.6
  • 189
    • 0037064165 scopus 로고    scopus 로고
    • Subthalamic GAD gene therapy in a Parkinson's disease rat model
    • doi: 10.1126/science.1074549
    • Luo, J., Kaplitt, M. G., Fitzsimons, H. L., Zuzga, D. S., Liu, Y., Oshinsky, M. L., et al. (2002). Subthalamic GAD gene therapy in a Parkinson's disease rat model. Science 298, 425-429. doi: 10.1126/science.1074549.
    • (2002) Science , vol.298 , pp. 425-429
    • Luo, J.1    Kaplitt, M.G.2    Fitzsimons, H.L.3    Zuzga, D.S.4    Liu, Y.5    Oshinsky, M.L.6
  • 190
    • 17844365885 scopus 로고    scopus 로고
    • Symptoms of autonomic failure in Parkinson's disease: prevalence and impact on daily life
    • doi: 10.1007/s10286-005-0253-z
    • Magerkurth, C., Schnitzer, R., and Braune, S. (2005). Symptoms of autonomic failure in Parkinson's disease: prevalence and impact on daily life. Clin. Auton. Res. 15, 76-82. doi: 10.1007/s10286-005-0253-z.
    • (2005) Clin. Auton. Res. , vol.15 , pp. 76-82
    • Magerkurth, C.1    Schnitzer, R.2    Braune, S.3
  • 191
    • 0030610127 scopus 로고    scopus 로고
    • Adenosine A2 receptors modulate haloperidol-induced catalepsy in rats
    • doi: 10.1016/S0014-2999(97)83039-7
    • Mandhane, S. N., Chopde, C. T., and Ghosh, A. K. (1997). Adenosine A2 receptors modulate haloperidol-induced catalepsy in rats. Eur. J. Pharmacol. 328, 135-141. doi: 10.1016/S0014-2999(97)83039-7.
    • (1997) Eur. J. Pharmacol. , vol.328 , pp. 135-141
    • Mandhane, S.N.1    Chopde, C.T.2    Ghosh, A.K.3
  • 192
    • 0030583640 scopus 로고    scopus 로고
    • MK-801 prevents levodopa-induced motor response alterations in parkinsonian rats
    • doi: 10.1016/0006-8993(96)00693-2
    • Marin, C., Papa, S., Engber, T. M., Bonastre, M., Tolosa, E., and Chase, T. N. (1996). MK-801 prevents levodopa-induced motor response alterations in parkinsonian rats. Brain Res. 736, 202-205. doi: 10.1016/0006-8993(96)00693-2.
    • (1996) Brain Res. , vol.736 , pp. 202-205
    • Marin, C.1    Papa, S.2    Engber, T.M.3    Bonastre, M.4    Tolosa, E.5    Chase, T.N.6
  • 193
    • 67650432423 scopus 로고    scopus 로고
    • Relationship between microglial activation and dopaminergic neuronal loss in the substantia nigra: a time course study in a 6-hydroxydopamine model of Parkinson's disease
    • doi: 10.1111/j.1471-4159.2009.06189.x
    • Marinova-Mutafchieva, L., Sadeghian, M., Broom, L., Davis, J. B., Medhurst, A. D., and Dexter, D. T. (2009). Relationship between microglial activation and dopaminergic neuronal loss in the substantia nigra: a time course study in a 6-hydroxydopamine model of Parkinson's disease. J. Neurochem. 110, 966-975. doi: 10.1111/j.1471-4159.2009.06189.x.
    • (2009) J. Neurochem. , vol.110 , pp. 966-975
    • Marinova-Mutafchieva, L.1    Sadeghian, M.2    Broom, L.3    Davis, J.B.4    Medhurst, A.D.5    Dexter, D.T.6
  • 194
    • 78349247631 scopus 로고    scopus 로고
    • Gene delivery of AAV2-neurturin for Parkinson's disease: a double-blind, randomised, controlled trial
    • doi: 10.1016/S1474-4422(10)70254-4
    • Marks, W. J. Jr. Bartus, R. T., Siffert, J., Davis, C. S., Lozano, A., Boulis, N., et al. (2010). Gene delivery of AAV2-neurturin for Parkinson's disease: a double-blind, randomised, controlled trial. Lancet Neurol. 9, 1164-1172. doi: 10.1016/S1474-4422(10)70254-4.
    • (2010) Lancet Neurol. , vol.9 , pp. 1164-1172
    • Marks Jr., W.J.1    Bartus, R.T.2    Siffert, J.3    Davis, C.S.4    Lozano, A.5    Boulis, N.6
  • 195
    • 41949110690 scopus 로고    scopus 로고
    • Safety and tolerability of intraputaminal delivery of CERE-120 (adeno-associated virus serotype 2-neurturin) to patients with idiopathic Parkinson's disease: an open-label, phase I trial
    • doi: 10.1016/S1474-4422(08)70065-6
    • Marks, W. J. Jr. Ostrem, J. L., Verhagen, L., Starr, P. A., Larson, P. S., Bakay, R. A., et al. (2008). Safety and tolerability of intraputaminal delivery of CERE-120 (adeno-associated virus serotype 2-neurturin) to patients with idiopathic Parkinson's disease: an open-label, phase I trial. Lancet Neurol. 7, 400-408. doi: 10.1016/S1474-4422(08)70065-6.
    • (2008) Lancet Neurol. , vol.7 , pp. 400-408
    • Marks Jr., W.J.1    Ostrem, J.L.2    Verhagen, L.3    Starr, P.A.4    Larson, P.S.5    Bakay, R.A.6
  • 196
    • 84863911386 scopus 로고    scopus 로고
    • A systematic review of catechol-0-methyltransferase inhibitors: efficacy and safety in clinical practice
    • doi: 10.1097/WNF.0b013e31825c034a
    • Marsala, S. Z., Gioulis, M., Ceravolo, R., and Tinazzi, M. (2012). A systematic review of catechol-0-methyltransferase inhibitors: efficacy and safety in clinical practice. Clin. Neuropharmacol. 35, 185-190. doi: 10.1097/WNF.0b013e31825c034a.
    • (2012) Clin. Neuropharmacol. , vol.35 , pp. 185-190
    • Marsala, S.Z.1    Gioulis, M.2    Ceravolo, R.3    Tinazzi, M.4
  • 197
    • 67349106731 scopus 로고    scopus 로고
    • A novel transferrin/TfR2-mediated mitochondrial iron transport system is disrupted in Parkinson's disease
    • doi: 10.1016/j.nbd.2009.02.009
    • Mastroberardino, P. G., Hoffman, E. K., Horowitz, M. P., Betarbet, R., Taylor, G., Cheng, D., et al. (2009). A novel transferrin/TfR2-mediated mitochondrial iron transport system is disrupted in Parkinson's disease. Neurobiol. Dis. 34, 417-431. doi: 10.1016/j.nbd.2009.02.009.
    • (2009) Neurobiol. Dis. , vol.34 , pp. 417-431
    • Mastroberardino, P.G.1    Hoffman, E.K.2    Horowitz, M.P.3    Betarbet, R.4    Taylor, G.5    Cheng, D.6
  • 198
    • 0025853857 scopus 로고
    • Effects of locus coeruleus lesions on parkinsonian signs, striatal dopamine and substantia nigra cell loss after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine in monkeys: a possible role for the locus coeruleus in the progression of Parkinson's disease
    • doi: 10.1016/0306-4522(91)90345-O
    • Mavridis, M., Degryse, A. D., Lategan, A. J., Marien, M. R., and Colpaert, F. C. (1991). Effects of locus coeruleus lesions on parkinsonian signs, striatal dopamine and substantia nigra cell loss after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine in monkeys: a possible role for the locus coeruleus in the progression of Parkinson's disease. Neuroscience 41, 507-523. doi: 10.1016/0306-4522(91)90345-O.
    • (1991) Neuroscience , vol.41 , pp. 507-523
    • Mavridis, M.1    Degryse, A.D.2    Lategan, A.J.3    Marien, M.R.4    Colpaert, F.C.5
  • 199
    • 0030608332 scopus 로고    scopus 로고
    • The competitive NMDA receptor antagonist SDZ 220-581 reverses haloperidol-induced catalepsy in rats
    • doi: 10.1016/S0014-2999(96)00576-6
    • McAllister, K. H. (1996). The competitive NMDA receptor antagonist SDZ 220-581 reverses haloperidol-induced catalepsy in rats. Eur. J. Pharmacol. 314, 307-311. doi: 10.1016/S0014-2999(96)00576-6.
    • (1996) Eur. J. Pharmacol. , vol.314 , pp. 307-311
    • McAllister, K.H.1
  • 200
    • 0023789193 scopus 로고
    • Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson's and Alzheimer's disease brains
    • doi: 10.1212/WNL.38.8.1285
    • Mcgeer, P. L., Itagaki, S., Boyes, B. E., and Mcgeer, E. G. (1988). Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson's and Alzheimer's disease brains. Neurology 38, 1285-1291. doi: 10.1212/WNL.38.8.1285.
    • (1988) Neurology , vol.38 , pp. 1285-1291
    • Mcgeer, P.L.1    Itagaki, S.2    Boyes, B.E.3    Mcgeer, E.G.4
  • 201
    • 0242384667 scopus 로고    scopus 로고
    • Presence of reactive microglia in monkey substantia nigra years after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine administration
    • doi: 10.1002/ana.10728
    • Mcgeer, P. L., Schwab, C., Parent, A., and Doudet, D. (2003). Presence of reactive microglia in monkey substantia nigra years after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine administration. Ann. Neurol. 54, 599-604. doi: 10.1002/ana.10728.
    • (2003) Ann. Neurol. , vol.54 , pp. 599-604
    • Mcgeer, P.L.1    Schwab, C.2    Parent, A.3    Doudet, D.4
  • 202
    • 79151475168 scopus 로고    scopus 로고
    • Differential response of the central noradrenergic nervous system to the loss of locus coeruleus neurons in Parkinson's disease and Alzheimer's disease
    • doi: 10.1016/j.brainres.2010.12.015
    • Mcmillan, P. J., White, S. S., Franklin, A., Greenup, J. L., Leverenz, J. B., Raskind, M. A., et al. (2011). Differential response of the central noradrenergic nervous system to the loss of locus coeruleus neurons in Parkinson's disease and Alzheimer's disease. Brain Res. 1373, 240-252. doi: 10.1016/j.brainres.2010.12.015.
    • (2011) Brain Res. , vol.1373 , pp. 240-252
    • Mcmillan, P.J.1    White, S.S.2    Franklin, A.3    Greenup, J.L.4    Leverenz, J.B.5    Raskind, M.A.6
  • 203
    • 0042134789 scopus 로고    scopus 로고
    • Mirtazapine in L-dopa-induced dyskinesias
    • doi: 10.1097/00002826-200307000-00005
    • Meco, G., Fabrizio, E., Di Rezze, S., Alessandri, A., and Pratesi, L. (2003). Mirtazapine in L-dopa-induced dyskinesias. Clin. Neuropharmacol. 26, 179-181. doi: 10.1097/00002826-200307000-00005.
    • (2003) Clin. Neuropharmacol. , vol.26 , pp. 179-181
    • Meco, G.1    Fabrizio, E.2    Di Rezze, S.3    Alessandri, A.4    Pratesi, L.5
  • 204
    • 67651149481 scopus 로고    scopus 로고
    • Aripiprazole in L-dopa-induced dyskinesias: a one-year open-label pilot study
    • doi: 10.1007/s00702-009-0231-z
    • Meco, G., Stirpe, P., Edito, F., Purcaro, C., Valente, M., Bernardi, S., et al. (2009). Aripiprazole in L-dopa-induced dyskinesias: a one-year open-label pilot study. J. Neural. Transm. 116, 881-884. doi: 10.1007/s00702-009-0231-z.
    • (2009) J. Neural. Transm. , vol.116 , pp. 881-884
    • Meco, G.1    Stirpe, P.2    Edito, F.3    Purcaro, C.4    Valente, M.5    Bernardi, S.6
  • 205
    • 0025017052 scopus 로고
    • Role of N-methyl-D-aspartate (NMDA) receptors in experimental catalepsy in rats
    • doi: 10.1016/0024-3205(90)90054-U
    • Mehta, A. K., and Ticku, M. K. (1990). Role of N-methyl-D-aspartate (NMDA) receptors in experimental catalepsy in rats. Life Sci. 46, 37-42. doi: 10.1016/0024-3205(90)90054-U.
    • (1990) Life Sci. , vol.46 , pp. 37-42
    • Mehta, A.K.1    Ticku, M.K.2
  • 206
    • 33747591029 scopus 로고    scopus 로고
    • Parkinson's disease: a rethink of rodent models
    • doi: 10.1007/s00221-006-0461-3
    • Melrose, H. L., Lincoln, S. J., Tyndall, G. M., and Farrer, M. J. (2006). Parkinson's disease: a rethink of rodent models. Exp. Brain Res. 173, 196-204. doi: 10.1007/s00221-006-0461-3.
    • (2006) Exp. Brain Res. , vol.173 , pp. 196-204
    • Melrose, H.L.1    Lincoln, S.J.2    Tyndall, G.M.3    Farrer, M.J.4
  • 207
    • 84891025045 scopus 로고    scopus 로고
    • APP Processing in human pluripotent stem cell-derived neurons is resistant to NSAID-based gamma-secretase modulation
    • doi: 10.1016/j.stemcr.2013.10.011
    • Mertens, J., Stuber, K., Wunderlich, P., Ladewig, J., Kesavan, J. C., Vandenberghe, R., et al. (2013). APP Processing in human pluripotent stem cell-derived neurons is resistant to NSAID-based gamma-secretase modulation. Stem. Cell. Reports 1, 491-498. doi: 10.1016/j.stemcr.2013.10.011.
    • (2013) Stem. Cell. Reports , vol.1 , pp. 491-498
    • Mertens, J.1    Stuber, K.2    Wunderlich, P.3    Ladewig, J.4    Kesavan, J.C.5    Vandenberghe, R.6
  • 208
    • 84905042090 scopus 로고    scopus 로고
    • Drug that May Slow Parkinson's Progression Granted $23 Million from NIH for Phase III testing.
    • New York, NY: Michael J. Fox Foundation. Available online at:, [Accessed 4 April 2014]
    • Michael, J. Fox Foundation. (2014). Drug that May Slow Parkinson's Progression Granted $23 Million from NIH for Phase III testing. New York, NY: Michael J. Fox Foundation. Available online at: https://www.michaeljfox.org/foundation/news-detail.php?podcast-drug-that-may-slow-parkinson-progression-granted-23-million-from-nih-for-phase-iii [Accessed 4 April 2014].
    • (2014)
    • Fox Foundation, M.J.1
  • 209
    • 84904989794 scopus 로고    scopus 로고
    • Addex Adds it Up for Parkinson's Patients.
    • New York, NY: The Michael J. Fox Foundation
    • Michael, J. Fox Foundation. (2012). Addex Adds it Up for Parkinson's Patients. New York, NY: The Michael J. Fox Foundation.
    • (2012)
    • Fox Foundation, M.J.1
  • 210
    • 0032738677 scopus 로고    scopus 로고
    • The absence of reactive astrocytosis is indicative of a unique inflammatory process in Parkinson's disease
    • doi: 10.1016/S0306-4522(99)00455-8
    • Mirza, B., Hadberg, H., Thomsen, P., and Moos, T. (2000). The absence of reactive astrocytosis is indicative of a unique inflammatory process in Parkinson's disease. Neuroscience 95, 425-432. doi: 10.1016/S0306-4522(99)00455-8.
    • (2000) Neuroscience , vol.95 , pp. 425-432
    • Mirza, B.1    Hadberg, H.2    Thomsen, P.3    Moos, T.4
  • 211
    • 82055181845 scopus 로고    scopus 로고
    • Differential regional expression patterns of alpha-synuclein, TNF-alpha, and IL-1beta ; and variable status of dopaminergic neurotoxicity in mouse brain after Paraquat treatment.
    • doi: 10.1186/1742-2094-8-163
    • Mitra, S., Chakrabarti, N., and Bhattacharyya, A. (2011). Differential regional expression patterns of alpha-synuclein, TNF-alpha, and IL-1beta ; and variable status of dopaminergic neurotoxicity in mouse brain after Paraquat treatment. J. Neuroinflammation. 8:163. doi: 10.1186/1742-2094-8-163.
    • (2011) J. Neuroinflammation , vol.8 , pp. 163
    • Mitra, S.1    Chakrabarti, N.2    Bhattacharyya, A.3
  • 212
    • 84860130544 scopus 로고    scopus 로고
    • Long-term evaluation of a phase 1 study of AADC gene therapy for Parkinson's disease
    • doi: 10.1089/hum.2011.220
    • Mittermeyer, G., Christine, C. W., Rosenbluth, K. H., Baker, S. L., Starr, P., Larson, P., et al. (2014). Long-term evaluation of a phase 1 study of AADC gene therapy for Parkinson's disease. Hum. Gene. Ther. 23, 377-381. doi: 10.1089/hum.2011.220.
    • (2014) Hum. Gene. Ther. , vol.23 , pp. 377-381
    • Mittermeyer, G.1    Christine, C.W.2    Rosenbluth, K.H.3    Baker, S.L.4    Starr, P.5    Larson, P.6
  • 213
    • 80053346792 scopus 로고    scopus 로고
    • Evidence-based initiation of dopaminergic therapy in Parkinson's disease
    • doi: 10.1007/s00415-010-5718-x
    • Miyasaki, J. M. (2010). Evidence-based initiation of dopaminergic therapy in Parkinson's disease. J. Neurol. 257, S309-S313. doi: 10.1007/s00415-010-5718-x.
    • (2010) J. Neurol. , vol.257
    • Miyasaki, J.M.1
  • 214
    • 0030838127 scopus 로고    scopus 로고
    • Glial cell line-derived neurotrophic factor-levodopa interactions and reduction of side effects in parkinsonian monkeys
    • doi: 10.1002/ana.410420212
    • Miyoshi, Y., Zhang, Z., Ovadia, A., Lapchak, P. A., Collins, F., Hilt, D., et al. (1997). Glial cell line-derived neurotrophic factor-levodopa interactions and reduction of side effects in parkinsonian monkeys. Ann. Neurol. 42, 208-214. doi: 10.1002/ana.410420212.
    • (1997) Ann. Neurol. , vol.42 , pp. 208-214
    • Miyoshi, Y.1    Zhang, Z.2    Ovadia, A.3    Lapchak, P.A.4    Collins, F.5    Hilt, D.6
  • 215
    • 84881555931 scopus 로고    scopus 로고
    • Adenosine A2A receptor antagonist istradefylline reduces daily OFF time in Parkinson's disease
    • doi: 10.1002/mds.25418
    • Mizuno, Y., and Kondo, T. (2013). Adenosine A2A receptor antagonist istradefylline reduces daily OFF time in Parkinson's disease. Mov. Disord. 28, 1138-1141. doi: 10.1002/mds.25418.
    • (2013) Mov. Disord. , vol.28 , pp. 1138-1141
    • Mizuno, Y.1    Kondo, T.2
  • 216
    • 0028171374 scopus 로고
    • Interleukin-1 beta, interleukin-6, epidermal growth factor and transforming growth factor-alpha are elevated in the brain from parkinsonian patients
    • doi: 10.1016/0304-3940(94)90508-8
    • Mogi, M., Harada, M., Kondo, T., Riederer, P., Inagaki, H., Minami, M., et al. (1994). Interleukin-1 beta, interleukin-6, epidermal growth factor and transforming growth factor-alpha are elevated in the brain from parkinsonian patients. Neurosci. Lett. 180, 147-150. doi: 10.1016/0304-3940(94)90508-8.
    • (1994) Neurosci. Lett. , vol.180 , pp. 147-150
    • Mogi, M.1    Harada, M.2    Kondo, T.3    Riederer, P.4    Inagaki, H.5    Minami, M.6
  • 217
    • 84871077687 scopus 로고    scopus 로고
    • Total CSF alpha-synuclein is lower in de novo Parkinson patients than in healthy subjects
    • doi: 10.1016/j.neulet.2012.11.004
    • Mollenhauer, B., Trautmann, E., Taylor, P., Manninger, P., Sixel-Doring, F., Ebentheuer, J., et al. (2013). Total CSF alpha-synuclein is lower in de novo Parkinson patients than in healthy subjects. Neurosci. Lett. 532, 44-48. doi: 10.1016/j.neulet.2012.11.004.
    • (2013) Neurosci. Lett. , vol.532 , pp. 44-48
    • Mollenhauer, B.1    Trautmann, E.2    Taylor, P.3    Manninger, P.4    Sixel-Doring, F.5    Ebentheuer, J.6
  • 218
    • 84876100906 scopus 로고    scopus 로고
    • Memantine for axial signs in Parkinson's disease: a randomised, double-blind, placebo-controlled pilot study
    • doi: 10.1136/jnnp-2012-303182
    • Moreau, C., Delval, A., Tiffreau, V., Defebvre, L., Dujardin, K., Duhamel, A., et al. (2013). Memantine for axial signs in Parkinson's disease: a randomised, double-blind, placebo-controlled pilot study. J. Neurol. Neurosurg. Psychiatry. 84, 552-555. doi: 10.1136/jnnp-2012-303182.
    • (2013) J. Neurol. Neurosurg. Psychiatry. , vol.84 , pp. 552-555
    • Moreau, C.1    Delval, A.2    Tiffreau, V.3    Defebvre, L.4    Dujardin, K.5    Duhamel, A.6
  • 219
    • 84872169926 scopus 로고    scopus 로고
    • MPEP, an mGlu5 receptor antagonist, reduces the development of L-DOPA-induced motor complications in de novo parkinsonian monkeys: biochemical correlates.
    • doi: 10.1016/j.neuropharm.2012.07.036
    • Morin, N., Gregoire, L., Morissette, M., Desrayaud, S., Gomez-Mancilla, B., Gasparini, F., et al. (2013a). MPEP, an mGlu5 receptor antagonist, reduces the development of L-DOPA-induced motor complications in de novo parkinsonian monkeys: biochemical correlates. Neuropharmacology 66, 355-364. doi: 10.1016/j.neuropharm.2012.07.036.
    • (2013) Neuropharmacology , vol.66 , pp. 355-364
    • Morin, N.1    Gregoire, L.2    Morissette, M.3    Desrayaud, S.4    Gomez-Mancilla, B.5    Gasparini, F.6
  • 220
    • 84885088944 scopus 로고    scopus 로고
    • Chronic treatment with MPEP, an mGlu5 receptor antagonist, normalizes basal ganglia glutamate neurotransmission in l-DOPA-treated parkinsonian monkeys.
    • doi: 10.1016/j.neuropharm.2013.05.028
    • Morin, N., Morissette, M., Gregoire, L., Gomez-Mancilla, B., Gasparini, F., and Di Paolo, T. (2013b). Chronic treatment with MPEP, an mGlu5 receptor antagonist, normalizes basal ganglia glutamate neurotransmission in l-DOPA-treated parkinsonian monkeys. Neuropharmacology 73C, 216-231. doi: 10.1016/j.neuropharm.2013.05.028.
    • (2013) Neuropharmacology , vol.73 , Issue.C , pp. 216-231
    • Morin, N.1    Morissette, M.2    Gregoire, L.3    Gomez-Mancilla, B.4    Gasparini, F.5    Di Paolo, T.6
  • 221
    • 84884204132 scopus 로고    scopus 로고
    • Microglia: a new frontier for synaptic plasticity, learning and memory, and neurodegenerative disease research
    • doi: 10.1016/j.nlm.2013.07.002
    • Morris, G. P., Clark, I. A., Zinn, R., and Vissel, B. (2013). Microglia: a new frontier for synaptic plasticity, learning and memory, and neurodegenerative disease research. Neurobiol. Learn. Mem. 105, 40-53. doi: 10.1016/j.nlm.2013.07.002.
    • (2013) Neurobiol. Learn. Mem. , vol.105 , pp. 40-53
    • Morris, G.P.1    Clark, I.A.2    Zinn, R.3    Vissel, B.4
  • 222
    • 58149107401 scopus 로고    scopus 로고
    • Combined 5-HT1A and 5-HT1B receptor agonists for the treatment of L-DOPA-induced dyskinesia
    • doi: 10.1093/brain/awn235
    • Munoz, A., Li, Q., Gardoni, F., Marcello, E., Qin, C., Carlsson, T., et al. (2008). Combined 5-HT1A and 5-HT1B receptor agonists for the treatment of L-DOPA-induced dyskinesia. Brain 131, 3380-3394. doi: 10.1093/brain/awn235.
    • (2008) Brain , vol.131 , pp. 3380-3394
    • Munoz, A.1    Li, Q.2    Gardoni, F.3    Marcello, E.4    Qin, C.5    Carlsson, T.6
  • 223
    • 77956262279 scopus 로고    scopus 로고
    • A phase I study of aromatic L-amino acid decarboxylase gene therapy for Parkinson's disease
    • doi: 10.1038/mt.2010.135
    • Muramatsu, S., Fujimoto, K., Kato, S., Mizukami, H., Asari, S., Ikeguchi, K., et al. (2010). A phase I study of aromatic L-amino acid decarboxylase gene therapy for Parkinson's disease. Mol. Ther. 18, 1731-1735. doi: 10.1038/mt.2010.135.
    • (2010) Mol. Ther. , vol.18 , pp. 1731-1735
    • Muramatsu, S.1    Fujimoto, K.2    Kato, S.3    Mizukami, H.4    Asari, S.5    Ikeguchi, K.6
  • 224
    • 0033834442 scopus 로고    scopus 로고
    • Antiparkinsonian actions of ifenprodil in the MPTP-lesioned marmoset model of Parkinson's disease
    • doi: 10.1006/exnr.2000.7444
    • Nash, J. E., Fox, S. H., Henry, B., Hill, M. P., Peggs, D., Mcguire, S., et al. (2000). Antiparkinsonian actions of ifenprodil in the MPTP-lesioned marmoset model of Parkinson's disease. Exp. Neurol. 165, 136-142. doi: 10.1006/exnr.2000.7444.
    • (2000) Exp. Neurol. , vol.165 , pp. 136-142
    • Nash, J.E.1    Fox, S.H.2    Henry, B.3    Hill, M.P.4    Peggs, D.5    Mcguire, S.6
  • 225
    • 77949516412 scopus 로고    scopus 로고
    • Metabotropic glutamate receptors: physiology, pharmacology, and disease
    • doi: 10.1146/annurev.pharmtox.011008.145533
    • Niswender, C. M., and Conn, P. J. (2010). Metabotropic glutamate receptors: physiology, pharmacology, and disease. Annu. Rev. Pharmacol. Toxicol. 50, 295-322. doi: 10.1146/annurev.pharmtox.011008.145533.
    • (2010) Annu. Rev. Pharmacol. Toxicol. , vol.50 , pp. 295-322
    • Niswender, C.M.1    Conn, P.J.2
  • 226
    • 55249109506 scopus 로고    scopus 로고
    • Histamine H(3) receptor ligands modulate L-dopa-evoked behavioral responses and L-dopa derived extracellular dopamine in dopamine-denervated rat striatum
    • doi: 10.1007/BF03033506
    • Nowak, P., Bortel, A., Dabrowska, J., Biedka, I., Slomian, G., Roczniak, W., et al. (2008). Histamine H(3) receptor ligands modulate L-dopa-evoked behavioral responses and L-dopa derived extracellular dopamine in dopamine-denervated rat striatum. Neurotox. Res. 13, 231-240. doi: 10.1007/BF03033506.
    • (2008) Neurotox. Res. , vol.13 , pp. 231-240
    • Nowak, P.1    Bortel, A.2    Dabrowska, J.3    Biedka, I.4    Slomian, G.5    Roczniak, W.6
  • 227
    • 0037435511 scopus 로고    scopus 로고
    • Randomized, double-blind trial of glial cell line-derived neurotrophic factor (GDNF) in PD
    • doi: 10.1212/WNL.60.1.69
    • Nutt, J. G., Burchiel, K. J., Comella, C. L., Jankovic, J., Lang, A. E., Laws, E. R. Jr., et al. (2003). Randomized, double-blind trial of glial cell line-derived neurotrophic factor (GDNF) in PD. Neurology 60, 69-73. doi: 10.1212/WNL.60.1.69.
    • (2003) Neurology , vol.60 , pp. 69-73
    • Nutt, J.G.1    Burchiel, K.J.2    Comella, C.L.3    Jankovic, J.4    Lang, A.E.5    Laws Jr., E.R.6
  • 228
    • 0037199818 scopus 로고    scopus 로고
    • Dopaminergic neuroprotection and regeneration by neurturin assessed by using behavioral, biochemical and histochemical measurements in a model of progressive Parkinson's disease
    • doi: 10.1016/S0006-8993(02)02934-7
    • Oiwa, Y., Yoshimura, R., Nakai, K., and Itakura, T. (2002). Dopaminergic neuroprotection and regeneration by neurturin assessed by using behavioral, biochemical and histochemical measurements in a model of progressive Parkinson's disease. Brain Res. 947, 271-283. doi: 10.1016/S0006-8993(02)02934-7.
    • (2002) Brain Res. , vol.947 , pp. 271-283
    • Oiwa, Y.1    Yoshimura, R.2    Nakai, K.3    Itakura, T.4
  • 229
    • 2342450729 scopus 로고    scopus 로고
    • Multicenter, open-label, trial of sarizotan in Parkinson disease patients with levodopa-induced dyskinesias (the SPLENDID Study)
    • doi: 10.1097/00002826-200403000-00003
    • Olanow, C. W., Damier, P., Goetz, C. G., Mueller, T., Nutt, J., Rascol, O., et al. (2004). Multicenter, open-label, trial of sarizotan in Parkinson disease patients with levodopa-induced dyskinesias (the SPLENDID Study). Clin. Neuropharmacol. 27, 58-62. doi: 10.1097/00002826-200403000-00003.
    • (2004) Clin. Neuropharmacol. , vol.27 , pp. 58-62
    • Olanow, C.W.1    Damier, P.2    Goetz, C.G.3    Mueller, T.4    Nutt, J.5    Rascol, O.6
  • 230
    • 70349456475 scopus 로고    scopus 로고
    • A double-blind, delayed-start trial of rasagiline in Parkinson's disease
    • doi: 10.1056/NEJMoa0809335
    • Olanow, C. W., Rascol, O., Hauser, R., Feigin, P. D., Jankovic, J., Lang, A., et al. (2009). A double-blind, delayed-start trial of rasagiline in Parkinson's disease. N. Engl. J. Med. 361, 1268-1278. doi: 10.1056/NEJMoa0809335.
    • (2009) N. Engl. J. Med. , vol.361 , pp. 1268-1278
    • Olanow, C.W.1    Rascol, O.2    Hauser, R.3    Feigin, P.D.4    Jankovic, J.5    Lang, A.6
  • 231
    • 0038754178 scopus 로고    scopus 로고
    • Parkin mutations and susceptibility alleles in late-onset Parkinson's disease
    • doi: 10.1002/ana.10524
    • Oliveira, S. A., Scott, W. K., Martin, E. R., Nance, M. A., Watts, R. L., Hubble, et al. (2003). Parkin mutations and susceptibility alleles in late-onset Parkinson's disease. Ann. Neurol. 53, 624-629. doi: 10.1002/ana.10524.
    • (2003) Ann. Neurol. , vol.53 , pp. 624-629
    • Oliveira, S.A.1    Scott, W.K.2    Martin, E.R.3    Nance, M.A.4    Watts, R.L.5    Hubble6
  • 232
    • 84897076939 scopus 로고    scopus 로고
    • Long-term safety and tolerability of ProSavin, a lentiviral vector-based gene therapy for Parkinson's disease: a dose escalation, open-label, phase 1/2 trial
    • doi: 10.1016/S0140-6736(13)61939-X
    • Palfi, S., Gurruchaga, J. M., Ralph, G. S., Lepetit, H., Lavisse, S., Buttery, P. C., et al. (2014). Long-term safety and tolerability of ProSavin, a lentiviral vector-based gene therapy for Parkinson's disease: a dose escalation, open-label, phase 1/2 trial. Lancet 383, 1138-1146. doi: 10.1016/S0140-6736(13)61939-X.
    • (2014) Lancet , vol.383 , pp. 1138-1146
    • Palfi, S.1    Gurruchaga, J.M.2    Ralph, G.S.3    Lepetit, H.4    Lavisse, S.5    Buttery, P.C.6
  • 233
    • 0037096359 scopus 로고    scopus 로고
    • Lentivirally delivered glial cell line-derived neurotrophic factor increases the number of striatal dopaminergic neurons in primate models of nigrostriatal degeneration
    • Palfi, S., Leventhal, L., Chu, Y., Ma, S. Y., Emborg, M., Bakay, R., et al. (2002). Lentivirally delivered glial cell line-derived neurotrophic factor increases the number of striatal dopaminergic neurons in primate models of nigrostriatal degeneration. J. Neurosci. 22, 4942-4954..
    • (2002) J. Neurosci. , vol.22 , pp. 4942-4954
    • Palfi, S.1    Leventhal, L.2    Chu, Y.3    Ma, S.Y.4    Emborg, M.5    Bakay, R.6
  • 234
    • 84878254691 scopus 로고    scopus 로고
    • NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease
    • doi: 10.1038/nrn3504
    • Paoletti, P., Bellone, C., and Zhou, Q. (2013). NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease. Nat. Rev. Neurosci. 14, 383-400. doi: 10.1038/nrn3504.
    • (2013) Nat. Rev. Neurosci. , vol.14 , pp. 383-400
    • Paoletti, P.1    Bellone, C.2    Zhou, Q.3
  • 235
    • 33846920665 scopus 로고    scopus 로고
    • NMDA receptor subunits: function and pharmacology
    • doi: 10.1016/j.coph.2006.08.011
    • Paoletti, P., and Neyton, J. (2007). NMDA receptor subunits: function and pharmacology. Curr. Opin. Pharmacol. 7, 39-47. doi: 10.1016/j.coph.2006.08.011.
    • (2007) Curr. Opin. Pharmacol. , vol.7 , pp. 39-47
    • Paoletti, P.1    Neyton, J.2
  • 236
    • 0024456653 scopus 로고
    • Effect of deprenyl on the progression of disability in early Parkinson's disease
    • Parkinson Study Group., doi: 10.1056/NEJM198911163212004
    • Parkinson Study Group. (1989). Effect of deprenyl on the progression of disability in early Parkinson's disease. The Parkinson Study Group. N. Engl. J. Med. 321, 1364-1371. doi: 10.1056/NEJM198911163212004.
    • (1989) The Parkinson Study Group. N. Engl. J. Med. , vol.321 , pp. 1364-1371
  • 237
    • 0027530638 scopus 로고
    • Effects of tocopherol and deprenyl on the progression of disability in early Parkinson's disease
    • Parkinson Study Group., doi: 10.1056/NEJM199301213280305
    • Parkinson Study Group. (1993). Effects of tocopherol and deprenyl on the progression of disability in early Parkinson's disease. N. Engl. J. Med. 328, 176-183. doi: 10.1056/NEJM199301213280305.
    • (1993) N. Engl. J. Med. , vol.328 , pp. 176-183
  • 238
    • 2342655732 scopus 로고    scopus 로고
    • A controlled, randomized, delayed-start study of rasagiline in early Parkinson disease
    • Parkinson Study Group., doi: 10.1001/archneur.61.4.561
    • Parkinson Study Group. (2004). A controlled, randomized, delayed-start study of rasagiline in early Parkinson disease. Arch. Neurol. 61, 561-566. doi: 10.1001/archneur.61.4.561.
    • (2004) Arch. Neurol. , vol.61 , pp. 561-566
  • 239
    • 84887625913 scopus 로고    scopus 로고
    • Phase II safety, tolerability, dose selection study of isradipine as a potential disease-modifying intervention in early Parkinson's disease (STEADY-PD).
    • Parkinson Study Group, . doi: 10.1002/mds.25639
    • Parkinson Study Group (2013). Phase II safety, tolerability, and dose selection study of isradipine as a potential disease-modifying intervention in early Parkinson's disease (STEADY-PD). Mov. Disord. 28, 1823-1831. doi: 10.1002/mds.25639.
    • (2013) Mov. Disord. , vol.28 , pp. 1823-1831
  • 240
    • 41149141259 scopus 로고    scopus 로고
    • Applicability of current staging/categorization of alpha-synuclein pathology and their clinical relevance
    • doi: 10.1007/s00401-008-0346-6
    • Parkkinen, L., Pirttila, T., and Alafuzoff, I. (2008). Applicability of current staging/categorization of alpha-synuclein pathology and their clinical relevance. Acta. Neuropathol. 115, 399-407. doi: 10.1007/s00401-008-0346-6.
    • (2008) Acta. Neuropathol. , vol.115 , pp. 399-407
    • Parkkinen, L.1    Pirttila, T.2    Alafuzoff, I.3
  • 242
    • 77957830241 scopus 로고    scopus 로고
    • Dopamine receptor agonists for the treatment of early or advanced Parkinson's disease
    • doi: 10.2165/11537810-000000000-00000
    • Perez-Lloret, S., and Rascol, O. (2010). Dopamine receptor agonists for the treatment of early or advanced Parkinson's disease. CNS Drugs 24, 941-968. doi: 10.2165/11537810-000000000-00000.
    • (2010) CNS Drugs , vol.24 , pp. 941-968
    • Perez-Lloret, S.1    Rascol, O.2
  • 243
    • 21544468215 scopus 로고    scopus 로고
    • Homeostatic plasticity and NMDA receptor trafficking
    • doi: 10.1016/j.tins.2005.03.004
    • Perez-Otano, I., and Ehlers, M. D. (2005). Homeostatic plasticity and NMDA receptor trafficking. Trends. Neurosci. 28, 229-238. doi: 10.1016/j.tins.2005.03.004.
    • (2005) Trends. Neurosci. , vol.28 , pp. 229-238
    • Perez-Otano, I.1    Ehlers, M.D.2
  • 244
    • 0028897149 scopus 로고
    • 5-HT receptors: past, present and future
    • doi: 10.1016/0166-2236(95)80023-U
    • Peroutka, S. J. (1995). 5-HT receptors: past, present and future. Trends. Neurosci. 18, 68-69. doi: 10.1016/0166-2236(95)80023-U.
    • (1995) Trends. Neurosci. , vol.18 , pp. 68-69
    • Peroutka, S.J.1
  • 245
    • 0036721026 scopus 로고    scopus 로고
    • Protection and reversal of excitotoxic neuronal damage by glucagon-like peptide-1 and exendin-4.
    • doi: 10.1124/jpet.102.037481
    • Perry, T., Haughey, N. J., Mattson, M. P., Egan, J. M., and Greig, N. H. (2002a). Protection and reversal of excitotoxic neuronal damage by glucagon-like peptide-1 and exendin-4. J Pharmacol. Exp. Ther. 302, 881-888. doi: 10.1124/jpet.102.037481.
    • (2002) J Pharmacol. Exp. Ther. , vol.302 , pp. 881-888
    • Perry, T.1    Haughey, N.J.2    Mattson, M.P.3    Egan, J.M.4    Greig, N.H.5
  • 246
    • 0036182251 scopus 로고    scopus 로고
    • A novel neurotrophic property of glucagon-like peptide 1: a promoter of nerve growth factor-mediated differentiation in PC12 cells.
    • doi: 10.1124/jpet.300.3.958
    • Perry, T., Lahiri, D. K., Chen, D., Zhou, J., Shaw, K. T., Egan, J. M., et al. (2002b). A novel neurotrophic property of glucagon-like peptide 1: a promoter of nerve growth factor-mediated differentiation in PC12 cells. J. Pharmacol. Exp. Ther. 300, 958-966. doi: 10.1124/jpet.300.3.958.
    • (2002) J. Pharmacol. Exp. Ther. , vol.300 , pp. 958-966
    • Perry, T.1    Lahiri, D.K.2    Chen, D.3    Zhou, J.4    Shaw, K.T.5    Egan, J.M.6
  • 247
    • 84858650685 scopus 로고    scopus 로고
    • Neurodegeneration and inflammation in Parkinson's disease
    • doi: 10.1016/S1353-8020(11)70064-5
    • Phani, S., Loike, J. D., and Przedborski, S. (2012). Neurodegeneration and inflammation in Parkinson's disease. Parkinsonism. Relat. Disord. 18(Suppl. 1), S207-S209. doi: 10.1016/S1353-8020(11)70064-5.
    • (2012) Parkinsonism. Relat. Disord. , vol.18 , Issue.SUPPL. 1
    • Phani, S.1    Loike, J.D.2    Przedborski, S.3
  • 248
    • 33646689597 scopus 로고    scopus 로고
    • Enhanced sensitivity of dopaminergic neurons to rotenone-induced toxicity with aging
    • doi: 10.1016/j.parkreldis.2005.12.002
    • Phinney, A. L., Andringa, G., Bol, J. G., Wolters, E., Van Muiswinkel, F. L., Van Dam, A. M., et al. (2006). Enhanced sensitivity of dopaminergic neurons to rotenone-induced toxicity with aging. Parkinsonism. Relat. Disord. 12, 228-238. doi: 10.1016/j.parkreldis.2005.12.002.
    • (2006) Parkinsonism. Relat. Disord. , vol.12 , pp. 228-238
    • Phinney, A.L.1    Andringa, G.2    Bol, J.G.3    Wolters, E.4    Van Muiswinkel, F.L.5    Van Dam, A.M.6
  • 249
    • 0028052238 scopus 로고
    • Polysynaptic regulation of glutamate receptors and mitochondrial enzyme activities in the basal ganglia of rats with unilateral dopamine depletion
    • Porter, R. H., Greene, J. G., Higgins, D. S. Jr., and Greenamyre, J. T. (1994). Polysynaptic regulation of glutamate receptors and mitochondrial enzyme activities in the basal ganglia of rats with unilateral dopamine depletion. J. Neurosci. 14, 7192-7199..
    • (1994) J. Neurosci. , vol.14 , pp. 7192-7199
    • Porter, R.H.1    Greene, J.G.2    Higgins Jr., D.S.3    Greenamyre, J.T.4
  • 250
    • 79251561841 scopus 로고    scopus 로고
    • Limitations of animal models of Parkinson's disease.
    • doi: 10.4061/2011/658083
    • Potashkin, J. A., Blume, S. R., and Runkle, N. K. (2011). Limitations of animal models of Parkinson's disease. Parkinsons. Dis. 2011:658083. doi: 10.4061/2011/658083.
    • (2011) Parkinsons. Dis. , vol.2011 , pp. 658083
    • Potashkin, J.A.1    Blume, S.R.2    Runkle, N.K.3
  • 251
    • 78649516910 scopus 로고    scopus 로고
    • Alterations in mGluR5 expression signaling in Lewy body disease and in transgenic models of alpha-synucleinopathy-implications for excitotoxicity.
    • doi: 10.1371/journal.pone.0014020
    • Price, D. L., Rockenstein, E., Ubhi, K., Phung, V., Maclean-Lewis, N., Askay, D., et al. (2010). Alterations in mGluR5 expression and signaling in Lewy body disease and in transgenic models of alpha-synucleinopathy-implications for excitotoxicity. PLoS ONE 5:e14020. doi: 10.1371/journal.pone.0014020.
    • (2010) PLoS ONE , vol.5
    • Price, D.L.1    Rockenstein, E.2    Ubhi, K.3    Phung, V.4    Maclean-Lewis, N.5    Askay, D.6
  • 252
    • 10244222286 scopus 로고    scopus 로고
    • MPTP as a mitochondrial neurotoxic model of Parkinson's disease
    • doi: 10.1023/B:JOBB.0000041771.66775.d5
    • Przedborski, S., Tieu, K., Perier, C., and Vila, M. (2004). MPTP as a mitochondrial neurotoxic model of Parkinson's disease. J. Bioenerg. Biomembr. 36, 375-379. doi: 10.1023/B:JOBB.0000041771.66775.d5.
    • (2004) J. Bioenerg. Biomembr. , vol.36 , pp. 375-379
    • Przedborski, S.1    Tieu, K.2    Perier, C.3    Vila, M.4
  • 253
    • 0037713489 scopus 로고    scopus 로고
    • The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model: a tool to explore the pathogenesis of Parkinson's disease
    • doi: 10.1111/j.1749-6632.2003.tb07476.x
    • Przedborski, S., and Vila, M. (2003). The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model: a tool to explore the pathogenesis of Parkinson's disease. Ann. N.Y. Acad. Sci. 991, 189-198. doi: 10.1111/j.1749-6632.2003.tb07476.x.
    • (2003) Ann. N.Y. Acad. Sci. , vol.991 , pp. 189-198
    • Przedborski, S.1    Vila, M.2
  • 254
    • 0035412888 scopus 로고    scopus 로고
    • Idazoxan, an alpha-2 antagonist, and L-DOPA-induced dyskinesias in patients with Parkinson's disease
    • doi: 10.1002/mds.1143
    • Rascol, O., Arnulf, I., Peyro-Saint Paul, H., Brefel-Courbon, C., Vidailhet, M., Thalamas, C., et al. (2001). Idazoxan, an alpha-2 antagonist, and L-DOPA-induced dyskinesias in patients with Parkinson's disease. Mov. Disord. 16, 708-713. doi: 10.1002/mds.1143.
    • (2001) Mov. Disord. , vol.16 , pp. 708-713
    • Rascol, O.1    Arnulf, I.2    Peyro-Saint Paul, H.3    Brefel-Courbon, C.4    Vidailhet, M.5    Thalamas, C.6
  • 255
    • 84855825060 scopus 로고    scopus 로고
    • Perampanel in Parkinson disease fluctuations: a double-blind randomized trial with placebo and entacapone
    • doi: 10.1097/WNF.0b013e318241520b
    • Rascol, O., Barone, P., Behari, M., Emre, M., Giladi, N., Olanow, C. W., et al. (2012). Perampanel in Parkinson disease fluctuations: a double-blind randomized trial with placebo and entacapone. Clin. Neuropharmacol. 35, 15-20. doi: 10.1097/WNF.0b013e318241520b.
    • (2012) Clin. Neuropharmacol. , vol.35 , pp. 15-20
    • Rascol, O.1    Barone, P.2    Behari, M.3    Emre, M.4    Giladi, N.5    Olanow, C.W.6
  • 256
    • 56849086964 scopus 로고    scopus 로고
    • Peripheral cytokines profile in Parkinson's disease
    • doi: 10.1016/j.bbi.2008.07.003
    • Reale, M., Iarlori, C., Thomas, A., Gambi, D., Perfetti, B., Di Nicola, M., et al. (2009). Peripheral cytokines profile in Parkinson's disease. Brain Behav. Immun. 23, 55-63. doi: 10.1016/j.bbi.2008.07.003.
    • (2009) Brain Behav. Immun. , vol.23 , pp. 55-63
    • Reale, M.1    Iarlori, C.2    Thomas, A.3    Gambi, D.4    Perfetti, B.5    Di Nicola, M.6
  • 257
    • 77958465056 scopus 로고    scopus 로고
    • Goal-directed and habitual control in the basal ganglia: implications for Parkinson's disease
    • doi: 10.1038/nrn2915
    • Redgrave, P., Rodriguez, M., Smith, Y., Rodriguez-Oroz, M. C., Lehericy, S., Bergman, H., et al. (2010). Goal-directed and habitual control in the basal ganglia: implications for Parkinson's disease. Nat. Rev. Neurosci. 11, 760-772. doi: 10.1038/nrn2915.
    • (2010) Nat. Rev. Neurosci. , vol.11 , pp. 760-772
    • Redgrave, P.1    Rodriguez, M.2    Smith, Y.3    Rodriguez-Oroz, M.C.4    Lehericy, S.5    Bergman, H.6
  • 258
    • 84860798931 scopus 로고    scopus 로고
    • Anti-hypertensive drugs as disease-modifying agents for Parkinson's disease: evidence from observational studies clinical trials. Cochrane.
    • doi: 10.1002/14651858.CD008535
    • Rees, K., Stowe, R., Patel, S., Ives, N., Breen, K., Ben-Shlomo, Y., et al. (2011). Anti-hypertensive drugs as disease-modifying agents for Parkinson's disease: evidence from observational studies and clinical trials. Cochrane. Database. Syst. Rev. 11:CD008535. doi: 10.1002/14651858.CD008535.
    • (2011) Database. Syst. Rev. , vol.11
    • Rees, K.1    Stowe, R.2    Patel, S.3    Ives, N.4    Breen, K.5    Ben-Shlomo, Y.6
  • 259
    • 84880006379 scopus 로고    scopus 로고
    • AAV2-mediated striatum delivery of human CDNF prevents the deterioration of midbrain dopamine neurons in a 6-hydroxydopamine induced parkinsonian rat model
    • doi: 10.1016/j.expneurol.2013.06.002
    • Ren, X., Zhang, T., Gong, X., Hu, G., Ding, W., and Wang, X. (2013). AAV2-mediated striatum delivery of human CDNF prevents the deterioration of midbrain dopamine neurons in a 6-hydroxydopamine induced parkinsonian rat model. Exp. Neurol. 248, 148-156. doi: 10.1016/j.expneurol.2013.06.002.
    • (2013) Exp. Neurol. , vol.248 , pp. 148-156
    • Ren, X.1    Zhang, T.2    Gong, X.3    Hu, G.4    Ding, W.5    Wang, X.6
  • 260
    • 0033044574 scopus 로고    scopus 로고
    • Can calcium antagonists provide a neuroprotective effect in Parkinson's disease?
    • doi: 10.2165/00003495-199957060-00001
    • Rodnitzky, R. L. (1999). Can calcium antagonists provide a neuroprotective effect in Parkinson's disease? Drugs 57, 845-849. doi: 10.2165/00003495-199957060-00001.
    • (1999) Drugs , vol.57 , pp. 845-849
    • Rodnitzky, R.L.1
  • 261
  • 262
    • 0032939002 scopus 로고    scopus 로고
    • Protection and regeneration of nigral dopaminergic neurons by neurturin or GDNF in a partial lesion model of Parkinson's disease after administration into the striatum or the lateral ventricle
    • doi: 10.1046/j.1460-9568.1999.00566.x
    • Rosenblad, C., Kirik, D., Devaux, B., Moffat, B., Phillips, H. S., and Bjorklund, A. (1999). Protection and regeneration of nigral dopaminergic neurons by neurturin or GDNF in a partial lesion model of Parkinson's disease after administration into the striatum or the lateral ventricle. Eur. J. Neurosci. 11, 1554-1566. doi: 10.1046/j.1460-9568.1999.00566.x.
    • (1999) Eur. J. Neurosci. , vol.11 , pp. 1554-1566
    • Rosenblad, C.1    Kirik, D.2    Devaux, B.3    Moffat, B.4    Phillips, H.S.5    Bjorklund, A.6
  • 263
    • 0142250855 scopus 로고    scopus 로고
    • Behavioral changes are not directly related to striatal monoamine levels, number of nigral neurons, or dose of parkinsonian toxin MPTP in mice
    • doi: 10.1016/S0969-9961(03)00108-6
    • Rousselet, E., Joubert, C., Callebert, J., Parain, K., Tremblay, L., Orieux, G., et al. (2003). Behavioral changes are not directly related to striatal monoamine levels, number of nigral neurons, or dose of parkinsonian toxin MPTP in mice. Neurobiol. Dis. 14, 218-228. doi: 10.1016/S0969-9961(03)00108-6.
    • (2003) Neurobiol. Dis. , vol.14 , pp. 218-228
    • Rousselet, E.1    Joubert, C.2    Callebert, J.3    Parain, K.4    Tremblay, L.5    Orieux, G.6
  • 264
    • 84882804747 scopus 로고    scopus 로고
    • Forebrain dopamine neurons project down to a brainstem region controlling locomotion
    • doi: 10.1073/pnas.1301125110
    • Ryczko, D., Gratsch, S., Auclair, F., Dube, C., Bergeron, S., Alpert, M. H., et al. (2013). Forebrain dopamine neurons project down to a brainstem region controlling locomotion. Proc. Natl. Acad. Sci. U.S.A. 110, E3235-E3242. doi: 10.1073/pnas.1301125110.
    • (2013) Proc. Natl. Acad. Sci. U.S.A. , vol.110
    • Ryczko, D.1    Gratsch, S.2    Auclair, F.3    Dube, C.4    Bergeron, S.5    Alpert, M.H.6
  • 265
    • 67649847847 scopus 로고    scopus 로고
    • Pharmacological modulation of glutamate transmission in a rat model of L-DOPA-induced dyskinesia: effects on motor behavior and striatal nuclear signaling
    • doi: 10.1124/jpet.108.150425
    • Rylander, D., Recchia, A., Mela, F., Dekundy, A., Danysz, W., and Cenci, M. A. (2009). Pharmacological modulation of glutamate transmission in a rat model of L-DOPA-induced dyskinesia: effects on motor behavior and striatal nuclear signaling. J. Pharmacol. Exp. Ther. 330, 227-235. doi: 10.1124/jpet.108.150425.
    • (2009) J. Pharmacol. Exp. Ther. , vol.330 , pp. 227-235
    • Rylander, D.1    Recchia, A.2    Mela, F.3    Dekundy, A.4    Danysz, W.5    Cenci, M.A.6
  • 266
    • 42649121783 scopus 로고    scopus 로고
    • Tremorolytic effects of adenosine A2A antagonists: implications for parkinsonism
    • doi: 10.2741/2952
    • Salamone, J. D., Betz, A. J., Ishiwari, K., Felsted, J., Madson, L., Mirante, B., et al. (2008). Tremorolytic effects of adenosine A2A antagonists: implications for parkinsonism. Front. Biosci. 13, 3594-3605. doi: 10.2741/2952.
    • (2008) Front. Biosci. , vol.13 , pp. 3594-3605
    • Salamone, J.D.1    Betz, A.J.2    Ishiwari, K.3    Felsted, J.4    Madson, L.5    Mirante, B.6
  • 267
    • 84892695519 scopus 로고    scopus 로고
    • Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer's disease
    • doi: 10.1056/NEJMoa1304839
    • Salloway, S., Sperling, R., Fox, N. C., Blennow, K., Klunk, W., Raskind, M., et al. (2014). Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer's disease. N. Engl. J. Med. 370, 322-333. doi: 10.1056/NEJMoa1304839.
    • (2014) N. Engl. J. Med. , vol.370 , pp. 322-333
    • Salloway, S.1    Sperling, R.2    Fox, N.C.3    Blennow, K.4    Klunk, W.5    Raskind, M.6
  • 268
    • 43849110886 scopus 로고    scopus 로고
    • mGluR5 metabotropic glutamate receptors and dyskinesias in MPTP monkeys
    • doi: 10.1016/j.neurobiolaging.2007.02.005
    • Samadi, P., Gregoire, L., Morissette, M., Calon, F., Hadj Tahar, A., Dridi, M., et al. (2008). mGluR5 metabotropic glutamate receptors and dyskinesias in MPTP monkeys. Neurobiol. Aging 29, 1040-1051. doi: 10.1016/j.neurobiolaging.2007.02.005.
    • (2008) Neurobiol. Aging , vol.29 , pp. 1040-1051
    • Samadi, P.1    Gregoire, L.2    Morissette, M.3    Calon, F.4    Hadj Tahar, A.5    Dridi, M.6
  • 269
    • 4344645719 scopus 로고    scopus 로고
    • Selective COX-2 inhibition prevents progressive dopamine neuron degeneration in a rat model of Parkinson's disease.
    • doi: 10.1186/1742-2094-1-6
    • Sanchez-Pernaute, R., Ferree, A., Cooper, O., Yu, M., Brownell, A. L., and Isacson, O. (2004). Selective COX-2 inhibition prevents progressive dopamine neuron degeneration in a rat model of Parkinson's disease. J. Neuroinflammation. 1:6. doi: 10.1186/1742-2094-1-6.
    • (2004) J. Neuroinflammation. , vol.1 , pp. 6
    • Sanchez-Pernaute, R.1    Ferree, A.2    Cooper, O.3    Yu, M.4    Brownell, A.L.5    Isacson, O.6
  • 270
    • 0034756359 scopus 로고    scopus 로고
    • Functional effect of adeno-associated virus mediated gene transfer of aromatic L-amino acid decarboxylase into the striatum of 6-OHDA-lesioned rats
    • doi: 10.1006/mthe.2001.0466
    • Sanchez-Pernaute, R., Harvey-White, J., Cunningham, J., and Bankiewicz, K. S. (2001). Functional effect of adeno-associated virus mediated gene transfer of aromatic L-amino acid decarboxylase into the striatum of 6-OHDA-lesioned rats. Mol. Ther. 4, 324-330. doi: 10.1006/mthe.2001.0466.
    • (2001) Mol. Ther. , vol.4 , pp. 324-330
    • Sanchez-Pernaute, R.1    Harvey-White, J.2    Cunningham, J.3    Bankiewicz, K.S.4
  • 271
    • 0029047329 scopus 로고
    • Glial cell line-derived neurotrophic factor but not transforming growth factor beta 3 prevents delayed degeneration of nigral dopaminergic neurons following striatal 6-hydroxydopamine lesion
    • doi: 10.1073/pnas.92.19.8935
    • Sauer, H., Rosenblad, C., and Bjorklund, A. (1995). Glial cell line-derived neurotrophic factor but not transforming growth factor beta 3 prevents delayed degeneration of nigral dopaminergic neurons following striatal 6-hydroxydopamine lesion. Proc. Natl. Acad. Sci. U.S.A. 92, 8935-8939. doi: 10.1073/pnas.92.19.8935.
    • (1995) Proc. Natl. Acad. Sci. U.S.A. , vol.92 , pp. 8935-8939
    • Sauer, H.1    Rosenblad, C.2    Bjorklund, A.3
  • 272
    • 0042474326 scopus 로고    scopus 로고
    • Fipamezole (JP-1730) is a potent alpha2 adrenergic receptor antagonist that reduces levodopa-induced dyskinesia in the MPTP-lesioned primate model of Parkinson's disease
    • doi: 10.1002/mds.10464
    • Savola, J. M., Hill, M., Engstrom, M., Merivuori, H., Wurster, S., McGuire, S. G., et al. (2003). Fipamezole (JP-1730) is a potent alpha2 adrenergic receptor antagonist that reduces levodopa-induced dyskinesia in the MPTP-lesioned primate model of Parkinson's disease. Mov. Disord. 18, 872-883. doi: 10.1002/mds.10464.
    • (2003) Mov. Disord. , vol.18 , pp. 872-883
    • Savola, J.M.1    Hill, M.2    Engstrom, M.3    Merivuori, H.4    Wurster, S.5    McGuire, S.G.6
  • 273
    • 82555196455 scopus 로고    scopus 로고
    • Monoamine oxidase B inhibitors for the treatment of Parkinson's disease: a review of symptomatic and potential disease-modifying effects
    • doi: 10.2165/11596310-000000000-00000
    • Schapira, A. H. (2011). Monoamine oxidase B inhibitors for the treatment of Parkinson's disease: a review of symptomatic and potential disease-modifying effects. CNS Drugs 25, 1061-1071. doi: 10.2165/11596310-000000000-00000.
    • (2011) CNS Drugs , vol.25 , pp. 1061-1071
    • Schapira, A.H.1
  • 274
    • 29244489587 scopus 로고    scopus 로고
    • Serotonergic mechanisms in Parkinson's disease: opposing results from preclinical and clinical data
    • doi: 10.1007/s00702-005-0368-3
    • Scholtissen, B., Verhey, F. R., Steinbusch, H. W., and Leentjens, A. F. (2006). Serotonergic mechanisms in Parkinson's disease: opposing results from preclinical and clinical data. J. Neural. Transm. 113, 59-73. doi: 10.1007/s00702-005-0368-3.
    • (2006) J. Neural. Transm. , vol.113 , pp. 59-73
    • Scholtissen, B.1    Verhey, F.R.2    Steinbusch, H.W.3    Leentjens, A.F.4
  • 275
    • 60349126440 scopus 로고    scopus 로고
    • Antagonizing L-type Ca2+ channel reduces development of abnormal involuntary movement in the rat model of L-3,4-dihydroxyphenylalanine-induced dyskinesia
    • doi: 10.1016/j.biopsych.2008.09.008
    • Schuster, S., Doudnikoff, E., Rylander, D., Berthet, A., Aubert, I., Ittrich, C., et al. (2009). Antagonizing L-type Ca2+ channel reduces development of abnormal involuntary movement in the rat model of L-3,4-dihydroxyphenylalanine-induced dyskinesia. Biol. Psychiatry. 65, 518-526. doi: 10.1016/j.biopsych.2008.09.008.
    • (2009) Biol. Psychiatry. , vol.65 , pp. 518-526
    • Schuster, S.1    Doudnikoff, E.2    Rylander, D.3    Berthet, A.4    Aubert, I.5    Ittrich, C.6
  • 276
    • 0346849912 scopus 로고    scopus 로고
    • Neuroprotection by a novel brain permeable iron chelator, VK-28, against 6-hydroxydopamine lession in rats
    • doi: 10.1016/j.neuropharm.2003.09.005
    • Shachar, D. B., Kahana, N., Kampel, V., Warshawsky, A., and Youdim, M. B. (2004). Neuroprotection by a novel brain permeable iron chelator, VK-28, against 6-hydroxydopamine lession in rats. Neuropharmacology 46, 254-263. doi: 10.1016/j.neuropharm.2003.09.005.
    • (2004) Neuropharmacology , vol.46 , pp. 254-263
    • Shachar, D.B.1    Kahana, N.2    Kampel, V.3    Warshawsky, A.4    Youdim, M.B.5
  • 277
    • 0037402801 scopus 로고    scopus 로고
    • Selective microglial activation in the rat rotenone model of Parkinson's disease
    • doi: 10.1016/S0304-3940(03)00172-1
    • Sherer, T. B., Betarbet, R., Kim, J. H., and Greenamyre, J. T. (2003). Selective microglial activation in the rat rotenone model of Parkinson's disease. Neurosci. Lett. 341, 87-90. doi: 10.1016/S0304-3940(03)00172-1.
    • (2003) Neurosci. Lett. , vol.341 , pp. 87-90
    • Sherer, T.B.1    Betarbet, R.2    Kim, J.H.3    Greenamyre, J.T.4
  • 278
    • 0033219932 scopus 로고    scopus 로고
    • Actions of adenosine A2A receptor antagonist KW-6002 on drug-induced catalepsy and hypokinesia caused by reserpine or MPTP
    • doi: 10.1007/s002130051146
    • Shiozaki, S., Ichikawa, S., Nakamura, J., Kitamura, S., Yamada, K., and Kuwana, Y. (1999). Actions of adenosine A2A receptor antagonist KW-6002 on drug-induced catalepsy and hypokinesia caused by reserpine or MPTP. Psychopharmacology (Berl.) 147, 90-95. doi: 10.1007/s002130051146.
    • (1999) Psychopharmacology (Berl.) , vol.147 , pp. 90-95
    • Shiozaki, S.1    Ichikawa, S.2    Nakamura, J.3    Kitamura, S.4    Yamada, K.5    Kuwana, Y.6
  • 279
    • 20344397157 scopus 로고    scopus 로고
    • Topiramate reduces levodopa-induced dyskinesia in the MPTP-lesioned marmoset model of Parkinson's disease
    • doi: 10.1002/mds.20345
    • Silverdale, M. A., Nicholson, S. L., Crossman, A. R., and Brotchie, J. M. (2005). Topiramate reduces levodopa-induced dyskinesia in the MPTP-lesioned marmoset model of Parkinson's disease. Mov. Disord. 20, 403-409. doi: 10.1002/mds.20345.
    • (2005) Mov. Disord. , vol.20 , pp. 403-409
    • Silverdale, M.A.1    Nicholson, S.L.2    Crossman, A.R.3    Brotchie, J.M.4
  • 280
    • 34548458484 scopus 로고    scopus 로고
    • The 6-hydroxydopamine model of Parkinson's disease
    • doi: 10.1007/BF03033565
    • Simola, N., Morelli, M., and Carta, A. R. (2007). The 6-hydroxydopamine model of Parkinson's disease. Neurotox. Res. 11, 151-167. doi: 10.1007/BF03033565.
    • (2007) Neurotox. Res. , vol.11 , pp. 151-167
    • Simola, N.1    Morelli, M.2    Carta, A.R.3
  • 281
    • 78650183301 scopus 로고    scopus 로고
    • Tolerability of isradipine in early Parkinson's disease: a pilot dose escalation study
    • doi: 10.1002/mds.23308
    • Simuni, T., Borushko, E., Avram, M. J., Miskevics, S., Martel, A., Zadikoff, C., et al. (2010). Tolerability of isradipine in early Parkinson's disease: a pilot dose escalation study. Mov. Disord. 25, 2863-2866. doi: 10.1002/mds.23308.
    • (2010) Mov. Disord. , vol.25 , pp. 2863-2866
    • Simuni, T.1    Borushko, E.2    Avram, M.J.3    Miskevics, S.4    Martel, A.5    Zadikoff, C.6
  • 282
    • 67651099082 scopus 로고    scopus 로고
    • Effect of caffeine on the expression of cytochrome P450 1A2, adenosine A2A receptor and dopamine transporter in control and 1-methyl 4-phenyl 1, 2, 3, 6-tetrahydropyridine treated mouse striatum
    • doi: 10.1016/j.brainres.2009.06.002
    • Singh, S., Singh, K., Gupta, S. P., Patel, D. K., Singh, V. K., Singh, R. K., et al. (2009). Effect of caffeine on the expression of cytochrome P450 1A2, adenosine A2A receptor and dopamine transporter in control and 1-methyl 4-phenyl 1, 2, 3, 6-tetrahydropyridine treated mouse striatum. Brain Res. 1283, 115-126. doi: 10.1016/j.brainres.2009.06.002.
    • (2009) Brain Res. , vol.1283 , pp. 115-126
    • Singh, S.1    Singh, K.2    Gupta, S.P.3    Patel, D.K.4    Singh, V.K.5    Singh, R.K.6
  • 283
    • 84873453232 scopus 로고    scopus 로고
    • The genetics of Parkinson's disease: progress and therapeutic implications
    • doi: 10.1002/mds.25249
    • Singleton, A. B., Farrer, M. J., and Bonifati, V. (2013). The genetics of Parkinson's disease: progress and therapeutic implications. Mov. Disord. 28, 14-23. doi: 10.1002/mds.25249.
    • (2013) Mov. Disord. , vol.28 , pp. 14-23
    • Singleton, A.B.1    Farrer, M.J.2    Bonifati, V.3
  • 284
    • 34047220707 scopus 로고    scopus 로고
    • Unilateral intraputamenal glial cell line-derived neurotrophic factor in patients with Parkinson disease: response to 1 year of treatment and 1 year of withdrawal
    • doi: 10.3171/jns.2007.106.4.614
    • Slevin, J. T., Gash, D. M., Smith, C. D., Gerhardt, G. A., Kryscio, R., Chebrolu, H., et al. (2007). Unilateral intraputamenal glial cell line-derived neurotrophic factor in patients with Parkinson disease: response to 1 year of treatment and 1 year of withdrawal. J. Neurosurg. 106, 614-620. doi: 10.3171/jns.2007.106.4.614.
    • (2007) J. Neurosurg. , vol.106 , pp. 614-620
    • Slevin, J.T.1    Gash, D.M.2    Smith, C.D.3    Gerhardt, G.A.4    Kryscio, R.5    Chebrolu, H.6
  • 285
    • 15744401358 scopus 로고    scopus 로고
    • The MPTP model of Parkinson's disease
    • doi: 10.1016/j.molbrainres.2004.09.017
    • Smeyne, R. J., and Jackson-Lewis, V. (2005). The MPTP model of Parkinson's disease. Brain Res. Mol. Brain Res. 134, 57-66. doi: 10.1016/j.molbrainres.2004.09.017.
    • (2005) Brain Res. Mol. Brain Res. , vol.134 , pp. 57-66
    • Smeyne, R.J.1    Jackson-Lewis, V.2
  • 286
    • 70349192980 scopus 로고    scopus 로고
    • Adenosine A2A receptor-antagonist/dopamine D2 receptor-agonist bivalent ligands as pharmacological tools to detect A2A-D2 receptor heteromers
    • doi: 10.1021/jm900298c
    • Soriano, A., Ventura, R., Molero, A., Hoen, R., Casado, V., Cortes, A., et al. (2009). Adenosine A2A receptor-antagonist/dopamine D2 receptor-agonist bivalent ligands as pharmacological tools to detect A2A-D2 receptor heteromers. J. Med. Chem. 52, 5590-5602. doi: 10.1021/jm900298c.
    • (2009) J. Med. Chem. , vol.52 , pp. 5590-5602
    • Soriano, A.1    Ventura, R.2    Molero, A.3    Hoen, R.4    Casado, V.5    Cortes, A.6
  • 287
    • 84878253190 scopus 로고    scopus 로고
    • Management of motor and non-motor symptoms in Parkinson's disease
    • doi: 10.1007/s40263-013-0053-2
    • Sprenger, F., and Poewe, W. (2013). Management of motor and non-motor symptoms in Parkinson's disease. CNS Drugs 27, 259-272. doi: 10.1007/s40263-013-0053-2.
    • (2013) CNS Drugs , vol.27 , pp. 259-272
    • Sprenger, F.1    Poewe, W.2
  • 288
    • 0027177628 scopus 로고
    • Blockade of 1-methyl-4-phenylpyridinium ion (MPP+) nigral toxicity in the rat by prior decortication or MK-801 treatment: a stereological estimate of neuronal loss
    • doi: 10.1016/0197-4580(93)90114-Q
    • Srivastava, R., Brouillet, E., Beal, M. F., Storey, E., and Hyman, B. T. (1993). Blockade of 1-methyl-4-phenylpyridinium ion (MPP+) nigral toxicity in the rat by prior decortication or MK-801 treatment: a stereological estimate of neuronal loss. Neurobiol. Aging. 14, 295-301. doi: 10.1016/0197-4580(93)90114-Q.
    • (1993) Neurobiol. Aging. , vol.14 , pp. 295-301
    • Srivastava, R.1    Brouillet, E.2    Beal, M.F.3    Storey, E.4    Hyman, B.T.5
  • 289
    • 0028813795 scopus 로고
    • Systemic administration of the NMDA receptor antagonist MK-801 potentiates circling induced by intrastriatal microinjection of dopamine
    • doi: 10.1016/0014-2999(94)00612-B
    • St-Pierre, J. A., and Bedard, P. J. (1995). Systemic administration of the NMDA receptor antagonist MK-801 potentiates circling induced by intrastriatal microinjection of dopamine. Eur. J. Pharmacol. 272, 123-129. doi: 10.1016/0014-2999(94)00612-B.
    • (1995) Eur. J. Pharmacol. , vol.272 , pp. 123-129
    • St-Pierre, J.A.1    Bedard, P.J.2
  • 290
    • 44949229417 scopus 로고    scopus 로고
    • A 12-week, placebo-controlled study (6002-US-006) of istradefylline in Parkinson disease
    • doi: 10.1212/01.wnl.0000313834.22171.17
    • Stacy, M., Silver, D., Mendis, T., Sutton, J., Mori, A., Chaikin, P., et al. (2008). A 12-week, placebo-controlled study (6002-US-006) of istradefylline in Parkinson disease. Neurology 70, 2233-2240. doi: 10.1212/01.wnl.0000313834.22171.17.
    • (2008) Neurology , vol.70 , pp. 2233-2240
    • Stacy, M.1    Silver, D.2    Mendis, T.3    Sutton, J.4    Mori, A.5    Chaikin, P.6
  • 291
    • 5444238860 scopus 로고    scopus 로고
    • Histological changes of the dopaminergic nigrostriatal system in aging
    • doi: 10.1007/s00441-004-0972-9
    • Stark, A. K., and Pakkenberg, B. (2004). Histological changes of the dopaminergic nigrostriatal system in aging. Cell. Tissue. Res. 318, 81-92. doi: 10.1007/s00441-004-0972-9.
    • (2004) Cell. Tissue. Res. , vol.318 , pp. 81-92
    • Stark, A.K.1    Pakkenberg, B.2
  • 292
    • 0034067617 scopus 로고    scopus 로고
    • Antiparkinsonian actions of CP-101,606, an antagonist of NR2B subunit-containing N-methyl-d-aspartate receptors
    • doi: 10.1006/exnr.2000.7374
    • Steece-Collier, K., Chambers, L. K., Jaw-Tsai, S. S., Menniti, F. S., and Greenamyre, J. T. (2000). Antiparkinsonian actions of CP-101,606, an antagonist of NR2B subunit-containing N-methyl-d-aspartate receptors. Exp. Neurol. 163, 239-243. doi: 10.1006/exnr.2000.7374.
    • (2000) Exp. Neurol. , vol.163 , pp. 239-243
    • Steece-Collier, K.1    Chambers, L.K.2    Jaw-Tsai, S.S.3    Menniti, F.S.4    Greenamyre, J.T.5
  • 293
    • 84887620872 scopus 로고    scopus 로고
    • AFQ056 in Parkinson patients with levodopa-induced dyskinesia: 13-week, randomized, dose-finding study
    • doi: 10.1002/mds.25561
    • Stocchi, F., Rascol, O., Destee, A., Hattori, N., Hauser, R. A., Lang, A. E., et al. (2013). AFQ056 in Parkinson patients with levodopa-induced dyskinesia: 13-week, randomized, dose-finding study. Mov. Disord. 28, 1838-1846. doi: 10.1002/mds.25561.
    • (2013) Mov. Disord. , vol.28 , pp. 1838-1846
    • Stocchi, F.1    Rascol, O.2    Destee, A.3    Hattori, N.4    Hauser, R.A.5    Lang, A.E.6
  • 295
    • 34247631275 scopus 로고    scopus 로고
    • Multiple hit hypotheses for dopamine neuron loss in Parkinson's disease
    • doi: 10.1016/j.tins.2007.03.009
    • Sulzer, D. (2007). Multiple hit hypotheses for dopamine neuron loss in Parkinson's disease. Trends. Neurosci. 30, 244-250. doi: 10.1016/j.tins.2007.03.009.
    • (2007) Trends. Neurosci. , vol.30 , pp. 244-250
    • Sulzer, D.1
  • 296
    • 0035134830 scopus 로고    scopus 로고
    • Inhibition of the cyclooxygenase isoenzymes COX-1 and COX-2 provide neuroprotection in the MPTP-mouse model of Parkinson's disease
    • doi: 10.1002/1098-2396(200102)39:2<167::AID-SYN8>3.0.CO;2-U
    • Teismann, P., and Ferger, B. (2001). Inhibition of the cyclooxygenase isoenzymes COX-1 and COX-2 provide neuroprotection in the MPTP-mouse model of Parkinson's disease. Synapse 39, 167-174. doi: 10.1002/1098-2396(200102)39:2<167::AID-SYN8>3.0.CO;2-U.
    • (2001) Synapse , vol.39 , pp. 167-174
    • Teismann, P.1    Ferger, B.2
  • 297
    • 33645894705 scopus 로고    scopus 로고
    • A randomized, double-blind, futility clinical trial of creatine and minocycline in early Parkinson disease
    • The Ninds Net-Pd Investigators., doi: 10.1212/01.wnl.0000201252.57661.e1
    • The Ninds Net-Pd Investigators. (2006). A randomized, double-blind, futility clinical trial of creatine and minocycline in early Parkinson disease. Neurology 66, 664-671. doi: 10.1212/01.wnl.0000201252.57661.e1.
    • (2006) Neurology , vol.66 , pp. 664-671
  • 298
    • 44649153832 scopus 로고    scopus 로고
    • A pilot clinical trial of creatine and minocycline in early Parkinson disease: 18-month results
    • The Ninds Net-Pd Investigators., doi: 10.1097/WNF.0b013e3181342f32
    • The Ninds Net-Pd Investigators. (2008). A pilot clinical trial of creatine and minocycline in early Parkinson disease: 18-month results. Clin. Neuropharmacol. 31, 141-150. doi: 10.1097/WNF.0b013e3181342f32.
    • (2008) Clin. Neuropharmacol. , vol.31 , pp. 141-150
  • 301
    • 0028834063 scopus 로고
    • Protection and repair of the nigrostriatal dopaminergic system by GDNF in vivo
    • doi: 10.1038/373335a0
    • Tomac, A., Lindqvist, E., Lin, L. F., Ogren, S. O., Young, D., Hoffer, B. J., et al. (1995). Protection and repair of the nigrostriatal dopaminergic system by GDNF in vivo. Nature 373, 335-339. doi: 10.1038/373335a0.
    • (1995) Nature , vol.373 , pp. 335-339
    • Tomac, A.1    Lindqvist, E.2    Lin, L.F.3    Ogren, S.O.4    Young, D.5    Hoffer, B.J.6
  • 302
    • 77952363301 scopus 로고    scopus 로고
    • Glutamate receptor ion channels: structure, regulation, and function
    • doi: 10.1124/pr.109.002451
    • Traynelis, S. F., Wollmuth, L. P., Mcbain, C. J., Menniti, F. S., Vance, K. M., Ogden, K. K., et al. (2010). Glutamate receptor ion channels: structure, regulation, and function. Pharmacol. Rev. 62, 405-496. doi: 10.1124/pr.109.002451.
    • (2010) Pharmacol. Rev. , vol.62 , pp. 405-496
    • Traynelis, S.F.1    Wollmuth, L.P.2    Mcbain, C.J.3    Menniti, F.S.4    Vance, K.M.5    Ogden, K.K.6
  • 303
    • 62249214963 scopus 로고    scopus 로고
    • Adenosine antagonists reverse the cataleptic effects of haloperidol: implications for the treatment of Parkinson's disease
    • doi: 10.1016/j.pbb.2009.02.001
    • Trevitt, J., Vallance, C., Harris, A., and Goode, T. (2009). Adenosine antagonists reverse the cataleptic effects of haloperidol: implications for the treatment of Parkinson's disease. Pharmacol. Biochem. Behav. 92, 521-527. doi: 10.1016/j.pbb.2009.02.001.
    • (2009) Pharmacol. Biochem. Behav. , vol.92 , pp. 521-527
    • Trevitt, J.1    Vallance, C.2    Harris, A.3    Goode, T.4
  • 304
    • 0026082060 scopus 로고
    • Protection of substantia nigra from MPP+ neurotoxicity by N-methyl-D-aspartate antagonists
    • doi: 10.1038/349414a0
    • Turski, L., Bressler, K., Rettig, K. J., Loschmann, P. A., and Wachtel, H. (1991). Protection of substantia nigra from MPP+ neurotoxicity by N-methyl-D-aspartate antagonists. Nature 349, 414-418. doi: 10.1038/349414a0.
    • (1991) Nature , vol.349 , pp. 414-418
    • Turski, L.1    Bressler, K.2    Rettig, K.J.3    Loschmann, P.A.4    Wachtel, H.5
  • 305
    • 77949495194 scopus 로고    scopus 로고
    • NMDA antagonist memantine improves levodopa-induced dyskinesias and "on-off" phenomena in Parkinson's disease
    • doi: 10.1002/mds.22917
    • Varanese, S., Howard, J., and Di Rocco, A. (2010). NMDA antagonist memantine improves levodopa-induced dyskinesias and "on-off" phenomena in Parkinson's disease. Mov. Disord. 25, 508-510. doi: 10.1002/mds.22917.
    • (2010) Mov. Disord. , vol.25 , pp. 508-510
    • Varanese, S.1    Howard, J.2    Di Rocco, A.3
  • 306
    • 76149098862 scopus 로고    scopus 로고
    • A2A adenosine receptor overexpression and functionality, as well as TNF-alpha levels, correlate with motor symptoms in Parkinson's disease
    • doi: 10.1096/fj.09-141044
    • Varani, K., Vincenzi, F., Tosi, A., Gessi, S., Casetta, I., Granieri, G., et al. (2010). A2A adenosine receptor overexpression and functionality, as well as TNF-alpha levels, correlate with motor symptoms in Parkinson's disease. FASEB J. 24, 587-598. doi: 10.1096/fj.09-141044.
    • (2010) FASEB J. , vol.24 , pp. 587-598
    • Varani, K.1    Vincenzi, F.2    Tosi, A.3    Gessi, S.4    Casetta, I.5    Granieri, G.6
  • 307
    • 0006647256 scopus 로고    scopus 로고
    • Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson's disease
    • doi: 10.1212/WNL.50.5.1323
    • Verhagen Metman, L., Del Dotto, P., Van Den Munckhof, P., Fang, J., Mouradian, M. M., and Chase, T. N. (1998). Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson's disease. Neurology 50, 1323-1326. doi: 10.1212/WNL.50.5.1323.
    • (1998) Neurology , vol.50 , pp. 1323-1326
    • Verhagen Metman, L.1    Del Dotto, P.2    Van Den Munckhof, P.3    Fang, J.4    Mouradian, M.M.5    Chase, T.N.6
  • 308
    • 26944465351 scopus 로고    scopus 로고
    • Neuroprotective effects of metabotropic glutamate receptor ligands in a 6-hydroxydopamine rodent model of Parkinson's disease
    • doi: 10.1111/j.1460-9568.2005.04362.x
    • Vernon, A. C., Palmer, S., Datla, K. P., Zbarsky, V., Croucher, M. J., and Dexter, D. T. (2005). Neuroprotective effects of metabotropic glutamate receptor ligands in a 6-hydroxydopamine rodent model of Parkinson's disease. Eur. J. Neurosci. 22, 1799-1806. doi: 10.1111/j.1460-9568.2005.04362.x.
    • (2005) Eur. J. Neurosci. , vol.22 , pp. 1799-1806
    • Vernon, A.C.1    Palmer, S.2    Datla, K.P.3    Zbarsky, V.4    Croucher, M.J.5    Dexter, D.T.6
  • 309
    • 35248859709 scopus 로고    scopus 로고
    • Subtype selective antagonism of substantia nigra pars compacta Group I metabotropic glutamate receptors protects the nigrostriatal system against 6-hydroxydopamine toxicity in vivo
    • doi: 10.1111/j.1471-4159.2007.04860.x
    • Vernon, A. C., Zbarsky, V., Datla, K. P., Croucher, M. J., and Dexter, D. T. (2007). Subtype selective antagonism of substantia nigra pars compacta Group I metabotropic glutamate receptors protects the nigrostriatal system against 6-hydroxydopamine toxicity in vivo. J. Neurochem. 103, 1075-1091. doi: 10.1111/j.1471-4159.2007.04860.x.
    • (2007) J. Neurochem. , vol.103 , pp. 1075-1091
    • Vernon, A.C.1    Zbarsky, V.2    Datla, K.P.3    Croucher, M.J.4    Dexter, D.T.5
  • 310
    • 0034986665 scopus 로고    scopus 로고
    • A use-dependent tyrosine dephosphorylation of NMDA receptors is independent of ion flux
    • doi: 10.1038/88404
    • Vissel, B., Krupp, J. J., Heinemann, S. F., and Westbrook, G. L. (2001). A use-dependent tyrosine dephosphorylation of NMDA receptors is independent of ion flux. Nat. Neurosci. 4, 587-596. doi: 10.1038/88404.
    • (2001) Nat. Neurosci. , vol.4 , pp. 587-596
    • Vissel, B.1    Krupp, J.J.2    Heinemann, S.F.3    Westbrook, G.L.4
  • 311
    • 0026668693 scopus 로고
    • NBQX (6-nitro-sulfamoyl-benzo-quinoxaline-dione) and CPP (3-carboxy-piperazin-propyl phosphonic acid) potentiate dopamine agonist induced rotations in substantia nigra lesioned rats
    • doi: 10.1016/0304-3940(92)90368-H
    • Wachtel, H., Kunow, M., and Loschmann, P. A. (1992). NBQX (6-nitro-sulfamoyl-benzo-quinoxaline-dione) and CPP (3-carboxy-piperazin-propyl phosphonic acid) potentiate dopamine agonist induced rotations in substantia nigra lesioned rats. Neurosci. Lett. 142, 179-182. doi: 10.1016/0304-3940(92)90368-H.
    • (1992) Neurosci. Lett. , vol.142 , pp. 179-182
    • Wachtel, H.1    Kunow, M.2    Loschmann, P.A.3
  • 312
    • 78649323061 scopus 로고    scopus 로고
    • Effect of 6-hydroxydopamine (6-OHDA) on proliferation of glial cells in the rat cortex and striatum: evidence for de-differentiation of resident astrocytes
    • doi: 10.1007/s00441-010-1061-x
    • Wachter, B., Schurger, S., Rolinger, J., Von Ameln-Mayerhofer, A., Berg, D., Wagner, H. J., et al. (2011). Effect of 6-hydroxydopamine (6-OHDA) on proliferation of glial cells in the rat cortex and striatum: evidence for de-differentiation of resident astrocytes. Cell. Tissue. Res. 342, 147-160. doi: 10.1007/s00441-010-1061-x.
    • (2011) Cell. Tissue. Res. , vol.342 , pp. 147-160
    • Wachter, B.1    Schurger, S.2    Rolinger, J.3    Von Ameln-Mayerhofer, A.4    Berg, D.5    Wagner, H.J.6
  • 313
    • 85047698430 scopus 로고    scopus 로고
    • Delayed delivery of AAV-GDNF prevents nigral neurodegeneration and promotes functional recovery in a rat model of Parkinson's disease
    • doi: 10.1038/sj.gt.3301682
    • Wang, L., Muramatsu, S., Lu, Y., Ikeguchi, K., Fujimoto, K., Okada, T., et al. (2002). Delayed delivery of AAV-GDNF prevents nigral neurodegeneration and promotes functional recovery in a rat model of Parkinson's disease. Gene. Ther. 9, 381-389. doi: 10.1038/sj.gt.3301682.
    • (2002) Gene. Ther. , vol.9 , pp. 381-389
    • Wang, L.1    Muramatsu, S.2    Lu, Y.3    Ikeguchi, K.4    Fujimoto, K.5    Okada, T.6
  • 314
    • 59149101888 scopus 로고    scopus 로고
    • Etiopathogenesis of Parkinson disease: a new beginning?
    • doi: 10.1177/1073858408319974
    • Weidong, L., Shen, C., and Jankovic, J. (2009). Etiopathogenesis of Parkinson disease: a new beginning? Neuroscientist 15, 28-35. doi: 10.1177/1073858408319974.
    • (2009) Neuroscientist , vol.15 , pp. 28-35
    • Weidong, L.1    Shen, C.2    Jankovic, J.3
  • 315
    • 0034118103 scopus 로고    scopus 로고
    • Coupled oscillator model of the dopaminergic neuron of the substantia nigra
    • Wilson, C. J., and Callaway, J. C. (2000). Coupled oscillator model of the dopaminergic neuron of the substantia nigra. J. Neurophysiol. 83, 3084-3100..
    • (2000) J. Neurophysiol. , vol.83 , pp. 3084-3100
    • Wilson, C.J.1    Callaway, J.C.2
  • 316
    • 77958569164 scopus 로고    scopus 로고
    • A role for calcium-permeable AMPA receptors in synaptic plasticity learning.
    • doi: 10.1371/journal.pone.0012818
    • Wiltgen, B. J., Royle, G. A., Gray, E. E., Abdipranoto, A., Thangthaeng, N., Jacobs, N., et al. (2010). A role for calcium-permeable AMPA receptors in synaptic plasticity and learning. PLoS ONE 5:12818. doi: 10.1371/journal.pone.0012818.
    • (2010) PLoS ONE , vol.5 , pp. 12818
    • Wiltgen, B.J.1    Royle, G.A.2    Gray, E.E.3    Abdipranoto, A.4    Thangthaeng, N.5    Jacobs, N.6
  • 317
    • 77953184604 scopus 로고    scopus 로고
    • Long-term antidyskinetic efficacy of amantadine in Parkinson's disease
    • doi: 10.1002/mds.23034
    • Wolf, E., Seppi, K., Katzenschlager, R., Hochschorner, G., Ransmayr, G., Schwingenschuh, P., et al. (2010). Long-term antidyskinetic efficacy of amantadine in Parkinson's disease. Mov. Disord. 25, 1357-1363. doi: 10.1002/mds.23034.
    • (2010) Mov. Disord. , vol.25 , pp. 1357-1363
    • Wolf, E.1    Seppi, K.2    Katzenschlager, R.3    Hochschorner, G.4    Ransmayr, G.5    Schwingenschuh, P.6
  • 318
    • 84863992209 scopus 로고    scopus 로고
    • The essential role of AMPA receptor GluR2 subunit RNA editing in the normal and diseased brain.
    • doi: 10.3389/fnmol.2012.00034
    • Wright, A., and Vissel, B. (2012). The essential role of AMPA receptor GluR2 subunit RNA editing in the normal and diseased brain. Front. Mol. Neurosci. 5:34. doi: 10.3389/fnmol.2012.00034.
    • (2012) Front. Mol. Neurosci. , vol.5 , pp. 34
    • Wright, A.1    Vissel, B.2
  • 319
    • 84864592590 scopus 로고    scopus 로고
    • Basal ganglia circuits changes in Parkinson's disease patients
    • doi: 10.1016/j.neulet.2012.07.012
    • Wu, T., Wang, J., Wang, C., Hallett, M., Zang, Y., Wu, X., et al. (2012). Basal ganglia circuits changes in Parkinson's disease patients. Neurosci. Lett. 524, 55-59. doi: 10.1016/j.neulet.2012.07.012.
    • (2012) Neurosci. Lett. , vol.524 , pp. 55-59
    • Wu, T.1    Wang, J.2    Wang, C.3    Hallett, M.4    Zang, Y.5    Wu, X.6
  • 320
    • 0028222272 scopus 로고
    • Glutamate receptors in striatum and substantia nigra: effects of medial forebrain bundle lesions
    • doi: 10.1016/0006-8993(94)91642-X
    • Wullner, U., Testa, C. M., Catania, M. V., Young, A. B., and Penney, J. B. Jr. (1994). Glutamate receptors in striatum and substantia nigra: effects of medial forebrain bundle lesions. Brain Res. 645, 98-102. doi: 10.1016/0006-8993(94)91642-X.
    • (1994) Brain Res. , vol.645 , pp. 98-102
    • Wullner, U.1    Testa, C.M.2    Catania, M.V.3    Young, A.B.4    Penney Jr., J.B.5
  • 321
    • 0037192439 scopus 로고    scopus 로고
    • Caffeine's neuroprotection against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxicity shows no tolerance to chronic caffeine administration in mice
    • doi: 10.1016/S0304-3940(02)00069-1
    • Xu, K., Xu, Y. H., Chen, J. F., and Schwarzschild, M. A. (2002). Caffeine's neuroprotection against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxicity shows no tolerance to chronic caffeine administration in mice. Neurosci. Lett. 322, 13-16. doi: 10.1016/S0304-3940(02)00069-1.
    • (2002) Neurosci. Lett. , vol.322 , pp. 13-16
    • Xu, K.1    Xu, Y.H.2    Chen, J.F.3    Schwarzschild, M.A.4
  • 322
    • 54349089328 scopus 로고    scopus 로고
    • The effects of MPTP on the activation of microglia/astrocytes and cytokine/chemokine levels in different mice strains
    • doi: 10.1016/j.jneuroim.2008.08.003
    • Yasuda, Y., Shimoda, T., Uno, K., Tateishi, N., Furuya, S., Yagi, K., et al. (2008). The effects of MPTP on the activation of microglia/astrocytes and cytokine/chemokine levels in different mice strains. J. Neuroimmunol. 204, 43-51. doi: 10.1016/j.jneuroim.2008.08.003.
    • (2008) J. Neuroimmunol. , vol.204 , pp. 43-51
    • Yasuda, Y.1    Shimoda, T.2    Uno, K.3    Tateishi, N.4    Furuya, S.5    Yagi, K.6
  • 323
    • 84858667840 scopus 로고    scopus 로고
    • M30, a brain permeable multitarget neurorestorative drug in post nigrostriatal dopamine neuron lesion of parkinsonism animal models
    • doi: 10.1016/S1353-8020(11)70047-5
    • Youdim, M. B. (2012). M30, a brain permeable multitarget neurorestorative drug in post nigrostriatal dopamine neuron lesion of parkinsonism animal models. Parkinsonism. Relat. Disord. 18(Suppl. 1), S151-S154. doi: 10.1016/S1353-8020(11)70047-5.
    • (2012) Parkinsonism. Relat. Disord. , vol.18 , Issue.SUPPL. 1
    • Youdim, M.B.1
  • 324
    • 1842608744 scopus 로고    scopus 로고
    • Ironing iron out in Parkinson's disease and other neurodegenerative diseases with iron chelators: a lesson from 6-hydroxydopamine and iron chelators, desferal and VK-28
    • doi: 10.1196/annals.1306.025
    • Youdim, M. B., Stephenson, G., and Ben Shachar, D. (2004). Ironing iron out in Parkinson's disease and other neurodegenerative diseases with iron chelators: a lesson from 6-hydroxydopamine and iron chelators, desferal and VK-28. Ann. N.Y. Acad. Sci. 1012, 306-325. doi: 10.1196/annals.1306.025.
    • (2004) Ann. N.Y. Acad. Sci. , vol.1012 , pp. 306-325
    • Youdim, M.B.1    Stephenson, G.2    Ben Shachar, D.3
  • 325
    • 0037337815 scopus 로고    scopus 로고
    • Neuronal loss is greater in the locus coeruleus than nucleus basalis and substantia nigra in Alzheimer and Parkinson diseases
    • doi: 10.1001/archneur.60.3.337
    • Zarow, C., Lyness, S. A., Mortimer, J. A., and Chui, H. C. (2003). Neuronal loss is greater in the locus coeruleus than nucleus basalis and substantia nigra in Alzheimer and Parkinson diseases. Arch. Neurol. 60, 337-341. doi: 10.1001/archneur.60.3.337.
    • (2003) Arch. Neurol. , vol.60 , pp. 337-341
    • Zarow, C.1    Lyness, S.A.2    Mortimer, J.A.3    Chui, H.C.4
  • 326
    • 0030967428 scopus 로고    scopus 로고
    • Dose response to intraventricular glial cell line-derived neurotrophic factor administration in parkinsonian monkeys
    • Zhang, Z., Miyoshi, Y., Lapchak, P. A., Collins, F., Hilt, D., Lebel, C., et al. (1997). Dose response to intraventricular glial cell line-derived neurotrophic factor administration in parkinsonian monkeys. J. Pharmacol. Exp. Ther. 282, 1396-1401..
    • (1997) J. Pharmacol. Exp. Ther. , vol.282 , pp. 1396-1401
    • Zhang, Z.1    Miyoshi, Y.2    Lapchak, P.A.3    Collins, F.4    Hilt, D.5    Lebel, C.6
  • 327
    • 0025895634 scopus 로고
    • Serotonergic sprouting is induced by dopamine-lesion in substantia nigra of adult rat brain
    • doi: 10.1016/0006-8993(91)90553-8
    • Zhou, F. C., Bledsoe, S., and Murphy, J. (1991). Serotonergic sprouting is induced by dopamine-lesion in substantia nigra of adult rat brain. Brain Res. 556, 108-116. doi: 10.1016/0006-8993(91)90553-8.
    • (1991) Brain Res. , vol.556 , pp. 108-116
    • Zhou, F.C.1    Bledsoe, S.2    Murphy, J.3
  • 328
    • 0026752794 scopus 로고
    • MK-801 prevents 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced parkinsonism in primates.
    • doi: 10.1111/j.1471-4159.1992.tb09429.x
    • Zuddas, A., Oberto, G., Vaglini, F., Fascetti, F., Fornai, F., and Corsini, G. U. (1992a). MK-801 prevents 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced parkinsonism in primates. J. Neurochem. 59, 733-739. doi: 10.1111/j.1471-4159.1992.tb09429.x.
    • (1992) J. Neurochem. , vol.59 , pp. 733-739
    • Zuddas, A.1    Oberto, G.2    Vaglini, F.3    Fascetti, F.4    Fornai, F.5    Corsini, G.U.6
  • 329
    • 0026782773 scopus 로고
    • Pharmacologic modulation of MPTP toxicity: MK 801 in prevention of dopaminergic cell death in monkeys and mice.
    • doi: 10.1111/j.1749-6632.1992.tb24553.x
    • Zuddas, A., Vaglini, F., Fornai, F., Fascetti, F., Saginario, A., and Corsini, G. U. (1992b). Pharmacologic modulation of MPTP toxicity: MK 801 in prevention of dopaminergic cell death in monkeys and mice. Ann. N.Y. Acad. Sci. 648, 268-271. doi: 10.1111/j.1749-6632.1992.tb24553.x
    • (1992) Ann. N.Y. Acad. Sci. , vol.648 , pp. 268-271
    • Zuddas, A.1    Vaglini, F.2    Fornai, F.3    Fascetti, F.4    Saginario, A.5    Corsini, G.U.6


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.